1
|
Egawa R, Yawo H, Kuba H. Activity-dependent refinement of axonal projections forms one-to-one connection pattern in the developing chick ciliary ganglion. Front Cell Neurosci 2025; 19:1560402. [PMID: 40271539 PMCID: PMC12014593 DOI: 10.3389/fncel.2025.1560402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/27/2025] [Indexed: 04/25/2025] Open
Abstract
Although it is well established that initially overproduced synaptic connections are extensively remodeled through activity-dependent competition for postsynaptic innervation, the mechanisms determining the final number of postsynaptic targets per axon remain unclear. Here, we investigated the morphology of individual axonal projections during development and the influence of neural activity in the chick ciliary ganglion (CG), a traditional model system for synapse maturation. By single-axon tracing combining Brainbow labeling and tissue clearing, we revealed that by embryonic day 14 (E14), hundreds of preganglionic axons each establish a one-to-one synaptic connection with single CG neurons via a calyx-type presynaptic terminal enveloping the soma of its postsynaptic target. This homogeneous connection pattern emerged through presynaptic terminal maturation from bouton-like to calyx-like morphology and concurrent axonal branch pruning starting around E10. The calyx maturation was retarded by the presynaptic expression of genetically encoded tools for silencing neuronal activity, enhanced tetanus neurotoxin light chain (eTeNT) or Kir2.1, demonstrating the activity-dependence of this morphological refinement. These findings suggest that some presynaptic mechanisms as well as synaptic competition would operate to restrict the number of postsynaptic targets innervated by each axon in the CG. Together with the easy accessibility to single-axon tracing, our results highlight the potential of the chick CG as a model for investigating the presynaptic factors underlying circuit remodeling.
Collapse
Affiliation(s)
- Ryo Egawa
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Hiromu Yawo
- Department of Developmental Biology and Neuroscience, Tohoku University Graduate School of Life Sciences, Sendai, Japan
| | - Hiroshi Kuba
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| |
Collapse
|
2
|
Piña Novo D, Gao M, Fischer R, Richevaux L, Yu J, Barrett JM, Shepherd GMG. Cortical dynamics in hand/forelimb S1 and M1 evoked by brief photostimulation of the mouse's hand. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.02.626335. [PMID: 39677687 PMCID: PMC11642753 DOI: 10.1101/2024.12.02.626335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Spiking activity along synaptic circuits linking primary somatosensory (S1) and motor (M1) areas is fundamental for sensorimotor integration in cortex. Circuits along the ascending somatosensory pathway through mouse hand/forelimb S1 and M1 were recently described in detail (Yamawaki et al., 2021). Here, we characterize the peripherally evoked spiking dynamics in these two cortical areas. Brief (5 ms) optogenetic photostimulation of the hand generated short (~25 ms) barrages of activity first in S1 (onset latency 15 ms) then M1 (10 ms later). The estimated propagation speed was 20-fold faster from hand to S1 than from S1 to M1. Amplitudes in M1 were strongly attenuated. Responses were typically triphasic, with suppression and rebound following the initial peak. Evoked activity in S1 was biased to middle layers, consistent with thalamocortical connectivity, while that in M1 was biased to upper layers, consistent with corticocortical connectivity. Parvalbumin (PV) inhibitory interneurons were involved in each phase, accounting for three quarters of the initial spikes generated in S1, and their selective photostimulation sufficed to evoke suppression and rebound in both S1 and M1. Partial silencing of S1 by PV activation during hand stimulation reduced the M1 sensory responses. Overall, these results characterize how evoked spiking activity propagates along the hand/forelimb transcortical loop, and illuminate how in vivo cortical dynamics relate to the underlying synaptic circuit organization in this system.
Collapse
Affiliation(s)
- Daniela Piña Novo
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Mang Gao
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Rita Fischer
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Louis Richevaux
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Jianing Yu
- School of Life Sciences, Peking University, Beijing 100871, China
| | - John M. Barrett
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Gordon M. G. Shepherd
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
3
|
Jackson J, Loughlin H, Looman C, Yu C. Pallidothalamic Circuit-Selective Manipulation Ameliorates Motor Symptoms in a Rat Model of Parkinsonian. J Neurosci 2025; 45:e0555242025. [PMID: 39837660 PMCID: PMC11905351 DOI: 10.1523/jneurosci.0555-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 12/09/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025] Open
Abstract
Deep brain stimulation (DBS) effectively treats motor symptoms of advanced Parkinson's disease (PD), with the globus pallidus interna (GPi) commonly targeted. However, its therapeutic mechanisms remain unclear. We employed optogenetic stimulation in the entopeduncular nucleus (EP), the rat homolog of GPi, in a unilateral 6-hydroxydopamine lesioned female Sprague Dawley rat model of PD. We quantified behavioral effects of optogenetic EP DBS on motor symptoms and conducted single-unit recordings in EP and ventral lateral motor thalamus (VL) to examine changes in neural activity. High-frequency optogenetic EP DBS (75, 100, 130 Hz) reduced ipsilateral turning and corrected forelimb stepping, while low-frequency stimulation (5 and 20 Hz) had no effect. EP and VL neurons exhibited mixed response during stimulation, with both increased and decreased firing. The average firing rate of all recorded neurons in the EP and VL significantly increased at 130 Hz but not at other frequencies. Beta-band oscillatory activity was reduced in most EP neurons across high frequencies (75, 100, 130 Hz), while reductions in beta-band oscillations in VL occurred only at 130 Hz. These findings suggest that the neural firing rates within EP and VL circuits were differentially modulated by EP DBS; they may not fully explain the frequency-dependent behavioral effect. Instead, high-frequency optogenetic EP DBS at 130 Hz may ameliorate parkinsonian motor symptoms by reducing abnormal oscillatory activity in the EP-VL circuits. This study underscores the therapeutic potential of circuit-specific modulation in the pallidothalamic pathway using optogenetic EP DBS to alleviate motor deficits in a PD rat model.
Collapse
Affiliation(s)
- Jacob Jackson
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan 49931
| | - Hannah Loughlin
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan 49931
| | - Chloe Looman
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan 49931
| | - Chunxiu Yu
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan 49931
| |
Collapse
|
4
|
Gonzalez-Burgos G, Miyamae T, Nishihata Y, Krimer OL, Wade K, Fish KN, Arion D, Cai ZL, Xue M, Stauffer WR, Lewis DA. Synaptic alterations in pyramidal cells following genetic manipulation of neuronal excitability in monkey prefrontal cortex. J Neurophysiol 2025; 133:399-413. [PMID: 39740351 DOI: 10.1152/jn.00326.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/16/2024] [Accepted: 12/21/2024] [Indexed: 01/02/2025] Open
Abstract
The primate dorsolateral prefrontal cortex (DLPFC) displays unique in vivo activity patterns, but how in vivo activity regulates DLPFC pyramidal neuron (PN) properties remains unclear. We assessed the effects of in vivo Kir2.1 overexpression, a genetic silencing tool, on synapses in monkey DLPFC PNs. We show for the first time that recombinant ion channel expression successfully modifies the excitability of primate cortex neurons, producing effects on synaptic properties apparently different from those in the rodent cortex.
Collapse
Affiliation(s)
| | - Takeaki Miyamae
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Yosuke Nishihata
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Olga L Krimer
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Kirsten Wade
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Kenneth N Fish
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Dominique Arion
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Zhao-Lin Cai
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas, United States
| | - Mingshan Xue
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| | - William R Stauffer
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
5
|
Carmi I, Zoabi S, Bittan AM, Kellner S, Oz S, Heinrich R, Berlin S. A genetically encoded secreted toxin potentiates synaptic NMDA receptors in hippocampal neurons and confers neuroprotection. PNAS NEXUS 2025; 4:pgaf041. [PMID: 39959712 PMCID: PMC11826341 DOI: 10.1093/pnasnexus/pgaf041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 01/27/2025] [Indexed: 02/18/2025]
Abstract
NMDA receptors (NMDARs) play essential roles in neuronal development, survival, and synaptic plasticity, to name a few. However, dysregulation in receptors' activity can lead to neuronal and synaptic damage, contributing to the development of various brain pathologies. Current pharmacological treatments targeting NMDARs remain limited, for instance due to insufficient receptor selectivity and poor spatial targeting. Genetic approaches hold promise to overcome some of these issues; however, require genetically encodable NMDAR-modulating peptides, which are scarce. Here, we explored NMDAR-selective peptide toxins from marine cone snails, which resulted in the necessary engineering of a posttranslational modification-free variant of Conantokin-P (naked Con-P). The naked form is essential for expression in mammalian cells. We systematically explored the naked variant and discovered that naked Con-P maintains its ability to inhibit GluN2B-containing receptors, but uniquely acquired the ability to potentiate GluN2A-containing synaptic receptors. We then engineered a secreted naked Con-P that readily enhances NMDAR-mediated synaptic events in primary hippocampal neurons, and mitigates neuronal damage induced by staurosporine. We therefore provide a genetically encodable, subtype selective, and secreted bimodulator of NMDARs. This new variant and approach should pave the way for the development of additional genetic tools, specifically tailored to target NMDARs within distinct cellular populations in the brain.
Collapse
Affiliation(s)
- Ido Carmi
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3525433, Israel
| | - Shaden Zoabi
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3525433, Israel
| | - Asaf M Bittan
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3525433, Israel
| | - Shai Kellner
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3525433, Israel
| | - Shimrit Oz
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3525433, Israel
| | - Ronit Heinrich
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3525433, Israel
| | - Shai Berlin
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3525433, Israel
| |
Collapse
|
6
|
Duan X, Zhang C, Wu Y, Ju J, Xu Z, Li X, Liu Y, Ohdah S, Constantin OM, Pan Y, Lu Z, Wang C, Chen X, Gee CE, Nagel G, Hou ST, Gao S, Song K. Suppression of epileptic seizures by transcranial activation of K +-selective channelrhodopsin. Nat Commun 2025; 16:559. [PMID: 39789018 PMCID: PMC11718177 DOI: 10.1038/s41467-025-55818-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025] Open
Abstract
Optogenetics is a valuable tool for studying the mechanisms of neurological diseases and is now being developed for therapeutic applications. In rodents and macaques, improved channelrhodopsins have been applied to achieve transcranial optogenetic stimulation. While transcranial photoexcitation of neurons has been achieved, noninvasive optogenetic inhibition for treating hyperexcitability-induced neurological disorders has remained elusive. There is a critical need for effective inhibitory optogenetic tools that are highly light-sensitive and capable of suppressing neuronal activity in deep brain tissue. In this study, we developed a highly sensitive moderately K+-selective channelrhodopsin (HcKCR1-hs) by molecular engineering of the recently discovered Hyphochytrium catenoides kalium (potassium) channelrhodopsin 1. Transcranial activation of HcKCR1-hs significantly prolongs the time to the first seizure, increases survival, and decreases seizure activity in several status epilepticus mouse models. Our approach for transcranial optogenetic inhibition of neural hyperactivity may be adapted for cell type-specific neuromodulation in both basic and preclinical settings.
Collapse
Affiliation(s)
- Xiaodong Duan
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| | - Chong Zhang
- Department of Neurophysiology, Institute of Physiology, University Würzburg, Würzburg, Germany
| | - Yujie Wu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Jun Ju
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Zhe Xu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Xuanyi Li
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Yao Liu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Schugofa Ohdah
- Institute for Synaptic Neuroscience, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Oana M Constantin
- Institute for Synaptic Neuroscience, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Yifan Pan
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Zhonghua Lu
- Research Center for Primate Neuromodulation and Neuroimaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Cheng Wang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaojing Chen
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Christine E Gee
- Institute for Synaptic Neuroscience, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Georg Nagel
- Department of Neurophysiology, Institute of Physiology, University Würzburg, Würzburg, Germany
| | - Sheng-Tao Hou
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| | - Shiqiang Gao
- Department of Neurophysiology, Institute of Physiology, University Würzburg, Würzburg, Germany.
| | - Kun Song
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, and Brain Research Center, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
7
|
Rentsch D, Bergs A, Shao J, Elvers N, Ruse C, Seidenthal M, Aoki I, Gottschalk A. Tools and methods for cell ablation and cell inhibition in Caenorhabditis elegans. Genetics 2025; 229:1-48. [PMID: 39110015 PMCID: PMC11708922 DOI: 10.1093/genetics/iyae119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/16/2024] [Indexed: 01/11/2025] Open
Abstract
To understand the function of cells such as neurons within an organism, it can be instrumental to inhibit cellular function, or to remove the cell (type) from the organism, and thus to observe the consequences on organismic and/or circuit function and animal behavior. A range of approaches and tools were developed and used over the past few decades that act either constitutively or acutely and reversibly, in systemic or local fashion. These approaches make use of either drugs or genetically encoded tools. Also, there are acutely acting inhibitory tools that require an exogenous trigger like light. Here, we give an overview of such methods developed and used in the nematode Caenorhabditis elegans.
Collapse
Affiliation(s)
- Dennis Rentsch
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| | - Amelie Bergs
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| | - Jiajie Shao
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| | - Nora Elvers
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| | - Christiane Ruse
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| | - Marius Seidenthal
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| | - Ichiro Aoki
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| | - Alexander Gottschalk
- Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue Strasse 9, D-60438 Frankfurt, Germany
| |
Collapse
|
8
|
Lee TY, Weissenberger Y, King AJ, Dahmen JC. Midbrain encodes sound detection behavior without auditory cortex. eLife 2024; 12:RP89950. [PMID: 39688376 DOI: 10.7554/elife.89950] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024] Open
Abstract
Hearing involves analyzing the physical attributes of sounds and integrating the results of this analysis with other sensory, cognitive, and motor variables in order to guide adaptive behavior. The auditory cortex is considered crucial for the integration of acoustic and contextual information and is thought to share the resulting representations with subcortical auditory structures via its vast descending projections. By imaging cellular activity in the corticorecipient shell of the inferior colliculus of mice engaged in a sound detection task, we show that the majority of neurons encode information beyond the physical attributes of the stimulus and that the animals' behavior can be decoded from the activity of those neurons with a high degree of accuracy. Surprisingly, this was also the case in mice in which auditory cortical input to the midbrain had been removed by bilateral cortical lesions. This illustrates that subcortical auditory structures have access to a wealth of non-acoustic information and can, independently of the auditory cortex, carry much richer neural representations than previously thought.
Collapse
Affiliation(s)
- Tai-Ying Lee
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Yves Weissenberger
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Andrew J King
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Johannes C Dahmen
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
9
|
Kim B, Kim HA, Woo J, Lee HJ, Kim TK, Min H, Lee CJ, Im HI. Striatal Cholinergic Interneurons Control Physical Nicotine Withdrawal via Muscarinic Receptor Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402274. [PMID: 39491887 DOI: 10.1002/advs.202402274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 07/26/2024] [Indexed: 11/05/2024]
Abstract
Striatal cholinergic interneurons (ChIs) provide acetylcholine tone to the striatum and govern motor functions. Nicotine withdrawal elicits physical symptoms that dysregulate motor behavior. Here, the role of striatal ChIs in physical nicotine withdrawal is investigated. Mice under RNAi-dependent genetic inhibition of striatal ChIs (ChIGI) by suppressing the sodium channel subunit NaV1.1, lessening action potential generation and activity-dependent acetylcholine release is first generated. ChIGI markedly reduced the somatic signs of nicotine withdrawal without affecting other nicotine-dependent or striatum-associated behaviors. Multielectrode array (MEA) recording revealed that ChIGI reversed ex vivo nicotine-induced alterations in the number of neural population spikes in the dorsal striatum. Notably, the drug repurposing strategy revealed that a clinically-approved antimuscarinic drug, procyclidine, fully mimicked the therapeutic electrophysiological effects of ChIGI. Furthermore, both ChIGI and procyclidine prevented the nicotine withdrawal-induced reduction in striatal dopamine release in vivo. Lastly, therapeutic intervention with procyclidine dose-dependently diminished the physical signs of nicotine withdrawal. The data demonstrated that the striatal ChIs are a critical substrate of physical nicotine withdrawal and that muscarinic antagonism holds therapeutic potential against nicotine withdrawal.
Collapse
Affiliation(s)
- Baeksun Kim
- Center for Brain Function, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea National University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Han Ah Kim
- Center for Brain Function, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea National University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Junsung Woo
- Center for Glia-Neuron Interaction, Brain Science Institute, KIST, Seoul, 02792, Republic of Korea
| | - Hyeon-Jeong Lee
- Doping Control Center, KIST, Seoul, 02792, Republic of Korea
| | - Tae Kyoo Kim
- Center for Brain Function, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hophil Min
- Division of Bio-Medical Science & Technology, KIST School, Korea National University of Science and Technology (UST), Seoul, 02792, Republic of Korea
- Doping Control Center, KIST, Seoul, 02792, Republic of Korea
| | - C Justin Lee
- Center for Glia-Neuron Interaction, Brain Science Institute, KIST, Seoul, 02792, Republic of Korea
| | - Heh-In Im
- Center for Brain Function, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea National University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| |
Collapse
|
10
|
Driscoll N, Antonini MJ, Cannon TM, Maretich P, Olaitan G, Van VDP, Nagao K, Sahasrabudhe A, Paniagua EV, Frey EJ, Kim YJ, Hunt S, Hummel M, Mupparaju S, Jasanoff A, Venton BJ, Anikeeva P. Multifunctional Neural Probes Enable Bidirectional Electrical, Optical, and Chemical Recording and Stimulation In Vivo. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2408154. [PMID: 39506430 DOI: 10.1002/adma.202408154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 10/28/2024] [Indexed: 11/08/2024]
Abstract
Recording and modulation of neuronal activity enables the study of brain function in health and disease. While translational neuroscience relies on electrical recording and modulation techniques, mechanistic studies in rodent models leverage genetic precision of optical methods, such as optogenetics and fluorescent indicator imaging. In addition to electrical signal transduction, neurons produce and receive diverse chemical signals which motivate tools to probe and modulate neurochemistry. Although the past decade has delivered a wealth of technologies for electrophysiology, optogenetics, chemical sensing, and optical recording, combining these modalities within a single platform remains challenging. This work leverages materials selection and convergence fiber drawing to permit neural recording, electrical stimulation, optogenetics, fiber photometry, drug and gene delivery, and voltammetric recording of neurotransmitters within individual fibers. Composed of polymers and non-magnetic carbon-based conductors, these fibers are compatible with magnetic resonance imaging, enabling concurrent stimulation and whole-brain monitoring. Their utility is demonstrated in studies of the mesolimbic reward pathway by interfacing with the ventral tegmental area and nucleus accumbens in mice and characterizing the neurophysiological effects of a stimulant drug. This study highlights the potential of these fibers to probe electrical, optical, and chemical signaling across multiple brain regions in both mechanistic and translational studies.
Collapse
Affiliation(s)
| | | | - Taylor M Cannon
- Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Pema Maretich
- Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | | | | | - Keisuke Nagao
- Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | | | | | - Ethan J Frey
- Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Ye Ji Kim
- Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Sydney Hunt
- Stanford University, Stanford, CA, 94305, USA
| | - Melissa Hummel
- Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Sanju Mupparaju
- Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Alan Jasanoff
- Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - B Jill Venton
- The University of Virginia, Charlottesville, VA, 22904, USA
| | - Polina Anikeeva
- Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
11
|
Kabanova A, Yang M, Logothetis NK, Eschenko O. Partial chemogenetic inhibition of the locus coeruleus due to heterogeneous transduction of noradrenergic neurons preserved auditory salience processing in wild-type rats. Eur J Neurosci 2024; 60:6237-6253. [PMID: 39349382 DOI: 10.1111/ejn.16550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 08/30/2024] [Accepted: 09/09/2024] [Indexed: 10/02/2024]
Abstract
The acoustic startle reflex (ASR) and prepulse inhibition of the ASR (PPI) assess the efficiency of salience processing, a fundamental brain function that is impaired in many psychiatric conditions. Both ASR and PPI depend on noradrenergic transmission, yet the modulatory role of the locus coeruleus (LC) remains controversial. Clonidine (0.05 mg/kg, i.p.), an alpha2-adrenoreceptor agonist, strongly reduced the ASR amplitude. In contrast, chemogenetic LC inhibition only mildly suppressed the ASR and did affect the PPI in virus-transduced rats. The canine adenovirus type 2 (CAV2)-based vector carrying a gene cassette for the expression of inhibitory receptors (hM4Di) and noradrenergic cell-specific promoter (PRSx8) had high cell-type specificity (94.4 ± 3.1%) but resulted in heterogeneous virus transduction of DbH-positive LC neurons (range: 9.2-94.4%). Clozapine-N-oxide (CNO; 1 mg/kg, i.p.), a hM4Di actuator, caused the firing cessation of hM4Di-expressing LC neurons, yet complete inhibition of the entire population of LC neurons was not achieved. Case-based immunohistochemistry revealed that virus injections distal (> 150 μm) to the LC core resulted in partial LC transduction, while proximal (< 50 μm) injections caused neuronal loss due to virus neurotoxicity. Neither the ASR nor PPI differed between the intact and virus-transduced rats. Our results suggest that a residual activity of virus-non-transduced LC neurons might have been sufficient for mediating an unaltered ASR and PPI. Our study highlights the importance of a case-based assessment of the virus efficiency, specificity, and neurotoxicity for targeted cell populations and of considering these factors when interpreting behavioral effects in experiments employing chemogenetic modulation.
Collapse
Affiliation(s)
- Anna Kabanova
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Mingyu Yang
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Nikos K Logothetis
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
- International Center for Primate Brain Research, Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Institute of Neuroscience (ION), Chinese Academy of Sciences, Shanghai, China
- Division of Imaging Science and Biomedical Engineering, University of Manchester, Manchester, UK
| | - Oxana Eschenko
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| |
Collapse
|
12
|
Wiest A, Maurer JJ, Weber F, Chung S. A hypothalamic circuit mechanism underlying the impact of stress on memory and sleep. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.17.618467. [PMID: 39463948 PMCID: PMC11507874 DOI: 10.1101/2024.10.17.618467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Stress profoundly affects sleep and memory processes. Stress impairs memory consolidation, and similarly, disruptions in sleep compromise memory functions. Yet, the neural circuits underlying stress-induced sleep and memory disturbances are still not fully understood. Here, we show that activation of CRHPVN neurons, similar to acute restraint stress, decreases sleep and impairs memory in a spatial object recognition task. Conversely, inhibiting CRHPVN neurons during stress reverses stress-induced memory deficits while slightly increasing the amount of sleep. We found that both stress and stimulation of CRHPVN neurons activate neurons in the lateral hypothalamus (LH), and that their projections to the LH are critical for mediating stress-induced memory deficits and sleep disruptions. Our results suggest a pivotal role for CRHPVN neuronal pathways in regulating the adverse effects of stress on memory and sleep, an important step towards improving sleep and ameliorating the cognitive deficits that occur in stress-related disorders.
Collapse
Affiliation(s)
- Alyssa Wiest
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Pharmacology Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John J. Maurer
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Pharmacology Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Franz Weber
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shinjae Chung
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
13
|
Selfe JS, Steyn TJS, Shorer EF, Burman RJ, Düsterwald KM, Kraitzick AZ, Abdelfattah AS, Schreiter ER, Newey SE, Akerman CJ, Raimondo JV. All-optical reporting of inhibitory receptor driving force in the nervous system. Nat Commun 2024; 15:8913. [PMID: 39414774 PMCID: PMC11484818 DOI: 10.1038/s41467-024-53074-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 09/30/2024] [Indexed: 10/18/2024] Open
Abstract
Ionic driving forces provide the net electromotive force for ion movement across receptors, channels, and transporters, and are a fundamental property of all cells. In the nervous system, fast synaptic inhibition is mediated by chloride permeable GABAA and glycine receptors, and single-cell intracellular recordings have been the only method for estimating driving forces across these receptors (DFGABAA). Here we present a tool for quantifying inhibitory receptor driving force named ORCHID: all-Optical Reporting of CHloride Ion Driving force. We demonstrate ORCHID's ability to provide accurate, high-throughput measurements of resting and dynamic DFGABAA from genetically targeted cell types over multiple timescales. ORCHID confirms theoretical predictions about the biophysical mechanisms that establish DFGABAA, reveals differences in DFGABAA between neurons and astrocytes, and affords the first in vivo measurements of intact DFGABAA. This work extends our understanding of inhibitory synaptic transmission and demonstrates the potential for all-optical methods to assess ionic driving forces.
Collapse
Affiliation(s)
- Joshua S Selfe
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Teresa J S Steyn
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Eran F Shorer
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Neurology, School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, United States of America
| | - Richard J Burman
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Kira M Düsterwald
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Gatsby Computational Neuroscience Unit, University College London, London, United Kingdom
| | - Ariel Z Kraitzick
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Ahmed S Abdelfattah
- Department of Neuroscience, Brown University, Providence, Rhode Island, United States of America
- Carney Institute for Brain Science, Brown University, Providence, Rhode Island, United States of America
| | - Eric R Schreiter
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, United States of America
| | - Sarah E Newey
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Colin J Akerman
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Joseph V Raimondo
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa.
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa.
- Wellcome Centre for Infectious Disease Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
14
|
Elleman AV, Milicic N, Williams DJ, Simko J, Liu CJ, Haynes AL, Ehrlich DE, Makinson CD, Du Bois J. Behavioral control through the direct, focal silencing of neuronal activity. Cell Chem Biol 2024; 31:1324-1335.e20. [PMID: 38729162 PMCID: PMC11260259 DOI: 10.1016/j.chembiol.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 01/02/2024] [Accepted: 04/09/2024] [Indexed: 05/12/2024]
Abstract
The ability to optically stimulate and inhibit neurons has revolutionized neuroscience research. Here, we present a direct, potent, user-friendly chemical approach for optically silencing neurons. We have rendered saxitoxin (STX), a naturally occurring paralytic agent, transiently inert through chemical protection with a previously undisclosed nitrobenzyl-derived photocleavable group. Exposing the caged toxin, STX-bpc, to a brief (5 ms) pulse of light effects rapid release of a potent STX derivative and transient, spatially precise blockade of voltage-gated sodium channels (NaVs). We demonstrate the efficacy of STX-bpc for parametrically manipulating action potentials in mammalian neurons and brain slice. Additionally, we show the effectiveness of this reagent for silencing neural activity by dissecting sensory-evoked swimming in larval zebrafish. Photo-uncaging of STX-bpc is a straightforward method for non-invasive, reversible, spatiotemporally precise neural silencing without the need for genetic access, thus removing barriers for comparative research.
Collapse
Affiliation(s)
- Anna V Elleman
- Department of Chemistry, Stanford University, 333 Campus Drive, Stanford, CA 94305, USA
| | - Nikola Milicic
- Department of Integrative Biology, University of Wisconsin-Madison, 121 Integrative Biology Research Building, 1117 W Johnson St, Madison, WI 53706, USA
| | - Damian J Williams
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, 710 W 168th St, New York, NY 10032, USA
| | - Jane Simko
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, 710 W 168th St, New York, NY 10032, USA
| | - Christine J Liu
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, 710 W 168th St, New York, NY 10032, USA; Department of Neuroscience, Columbia University, Jerome L. Greene Science Center, 3227 Broadway, MC 9872, New York, NY 10027, USA
| | - Allison L Haynes
- Department of Chemistry, Stanford University, 333 Campus Drive, Stanford, CA 94305, USA
| | - David E Ehrlich
- Department of Integrative Biology, University of Wisconsin-Madison, 121 Integrative Biology Research Building, 1117 W Johnson St, Madison, WI 53706, USA.
| | - Christopher D Makinson
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, 710 W 168th St, New York, NY 10032, USA; Department of Neuroscience, Columbia University, Jerome L. Greene Science Center, 3227 Broadway, MC 9872, New York, NY 10027, USA.
| | - J Du Bois
- Department of Chemistry, Stanford University, 333 Campus Drive, Stanford, CA 94305, USA.
| |
Collapse
|
15
|
Steinhardt CR, Mitchell DE, Cullen KE, Fridman GY. Pulsatile electrical stimulation creates predictable, correctable disruptions in neural firing. Nat Commun 2024; 15:5861. [PMID: 38997274 PMCID: PMC11245474 DOI: 10.1038/s41467-024-49900-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Electrical stimulation is a key tool in neuroscience, both in brain mapping studies and in many therapeutic applications such as cochlear, vestibular, and retinal neural implants. Due to safety considerations, stimulation is restricted to short biphasic pulses. Despite decades of research and development, neural implants lead to varying restoration of function in patients. In this study, we use computational modeling to provide an explanation for how pulsatile stimulation affects axonal channels and therefore leads to variability in restoration of neural responses. The phenomenological explanation is transformed into equations that predict induced firing rate as a function of pulse rate, pulse amplitude, and spontaneous firing rate. We show that these equations predict simulated responses to pulsatile stimulation with a variety of parameters as well as several features of experimentally recorded primate vestibular afferent responses to pulsatile stimulation. We then discuss the implications of these effects for improving clinical stimulation paradigms and electrical stimulation-based experiments.
Collapse
Affiliation(s)
- Cynthia R Steinhardt
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Center for Theoretical Neuroscience, Columbia University, New York, NY, USA.
| | - Diana E Mitchell
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Kathleen E Cullen
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Otolaryngology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Gene Y Fridman
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Otolaryngology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
16
|
Johnsen KA, Cruzado NA, Menard ZC, Willats AA, Charles AS, Markowitz JE, Rozell CJ. Bridging model and experiment in systems neuroscience with Cleo: the Closed-Loop, Electrophysiology, and Optophysiology simulation testbed. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.27.525963. [PMID: 39026717 PMCID: PMC11257437 DOI: 10.1101/2023.01.27.525963] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Systems neuroscience has experienced an explosion of new tools for reading and writing neural activity, enabling exciting new experiments such as all-optical or closed-loop control that effect powerful causal interventions. At the same time, improved computational models are capable of reproducing behavior and neural activity with increasing fidelity. Unfortunately, these advances have drastically increased the complexity of integrating different lines of research, resulting in the missed opportunities and untapped potential of suboptimal experiments. Experiment simulation can help bridge this gap, allowing model and experiment to better inform each other by providing a low-cost testbed for experiment design, model validation, and methods engineering. Specifically, this can be achieved by incorporating the simulation of the experimental interface into our models, but no existing tool integrates optogenetics, two-photon calcium imaging, electrode recording, and flexible closed-loop processing with neural population simulations. To address this need, we have developed Cleo: the Closed-Loop, Electrophysiology, and Optophysiology experiment simulation testbed. Cleo is a Python package enabling injection of recording and stimulation devices as well as closed-loop control with realistic latency into a Brian spiking neural network model. It is the only publicly available tool currently supporting two-photon and multi-opsin/wavelength optogenetics. To facilitate adoption and extension by the community, Cleo is open-source, modular, tested, and documented, and can export results to various data formats. Here we describe the design and features of Cleo, validate output of individual components and integrated experiments, and demonstrate its utility for advancing optogenetic techniques in prospective experiments using previously published systems neuroscience models.
Collapse
Affiliation(s)
- Kyle A. Johnsen
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | | | - Zachary C. Menard
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Adam A. Willats
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Adam S. Charles
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Jeffrey E. Markowitz
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | | |
Collapse
|
17
|
Wietek J, Nozownik A, Pulin M, Saraf-Sinik I, Matosevich N, Gowrishankar R, Gat A, Malan D, Brown BJ, Dine J, Imambocus BN, Levy R, Sauter K, Litvin A, Regev N, Subramaniam S, Abrera K, Summarli D, Goren EM, Mizrachi G, Bitton E, Benjamin A, Copits BA, Sasse P, Rost BR, Schmitz D, Bruchas MR, Soba P, Oren-Suissa M, Nir Y, Wiegert JS, Yizhar O. A bistable inhibitory optoGPCR for multiplexed optogenetic control of neural circuits. Nat Methods 2024; 21:1275-1287. [PMID: 38811857 PMCID: PMC11239505 DOI: 10.1038/s41592-024-02285-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 04/18/2024] [Indexed: 05/31/2024]
Abstract
Information is transmitted between brain regions through the release of neurotransmitters from long-range projecting axons. Understanding how the activity of such long-range connections contributes to behavior requires efficient methods for reversibly manipulating their function. Chemogenetic and optogenetic tools, acting through endogenous G-protein-coupled receptor pathways, can be used to modulate synaptic transmission, but existing tools are limited in sensitivity, spatiotemporal precision or spectral multiplexing capabilities. Here we systematically evaluated multiple bistable opsins for optogenetic applications and found that the Platynereis dumerilii ciliary opsin (PdCO) is an efficient, versatile, light-activated bistable G-protein-coupled receptor that can suppress synaptic transmission in mammalian neurons with high temporal precision in vivo. PdCO has useful biophysical properties that enable spectral multiplexing with other optogenetic actuators and reporters. We demonstrate that PdCO can be used to conduct reversible loss-of-function experiments in long-range projections of behaving animals, thereby enabling detailed synapse-specific functional circuit mapping.
Collapse
Affiliation(s)
- Jonas Wietek
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel.
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel.
- Neuroscience Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | - Adrianna Nozownik
- Center for Molecular Neurobiology, Hamburg, Germany
- Paris Brain Institute, Institut du Cerveau (ICM), CNRS UMR 7225, INSERM U1127, Sorbonne Université, Paris, France
| | - Mauro Pulin
- Center for Molecular Neurobiology, Hamburg, Germany
- Laboratory of Sensory Processing, Brain Mind Institute, Faculty of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Inbar Saraf-Sinik
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Noa Matosevich
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Raajaram Gowrishankar
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Excellence in the Neurobiology of Addiction, Pain and Emotion, University of Washington, Seattle, WA, USA
| | - Asaf Gat
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Daniela Malan
- Institut für Physiologie I, University of Bonn, Bonn, Germany
| | - Bobbie J Brown
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Julien Dine
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
- Boehringer Ingelheim Pharma GmbH & Co. KG; CNS Diseases, Biberach an der Riss, Germany
| | | | - Rivka Levy
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | | | - Anna Litvin
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Noa Regev
- Department of Physiology and Pharmacology, Tel Aviv University, Tel Aviv, Israel
| | - Suraj Subramaniam
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Khalid Abrera
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
| | - Dustin Summarli
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
| | - Eva Madeline Goren
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- University of Michigan, Ann Arbor, MI, USA
| | - Gili Mizrachi
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal Bitton
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Asaf Benjamin
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Bryan A Copits
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Philipp Sasse
- Institut für Physiologie I, University of Bonn, Bonn, Germany
| | - Benjamin R Rost
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Neuroscience Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dietmar Schmitz
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Neuroscience Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience, Berlin, Germany
- Einstein Center for Neurosciences, Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Michael R Bruchas
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Excellence in the Neurobiology of Addiction, Pain and Emotion, University of Washington, Seattle, WA, USA
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Peter Soba
- LIMES-Institute, University of Bonn, Bonn, Germany
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Meital Oren-Suissa
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Yuval Nir
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Department of Physiology and Pharmacology, Tel Aviv University, Tel Aviv, Israel
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - J Simon Wiegert
- Center for Molecular Neurobiology, Hamburg, Germany
- MCTN, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Ofer Yizhar
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel.
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
18
|
Maurer JJ, Lin A, Jin X, Hong J, Sathi N, Cardis R, Osorio-Forero A, Lüthi A, Weber F, Chung S. Homeostatic regulation of rapid eye movement sleep by the preoptic area of the hypothalamus. eLife 2024; 12:RP92095. [PMID: 38884573 PMCID: PMC11182646 DOI: 10.7554/elife.92095] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024] Open
Abstract
Rapid eye movement sleep (REMs) is characterized by activated electroencephalogram (EEG) and muscle atonia, accompanied by vivid dreams. REMs is homeostatically regulated, ensuring that any loss of REMs is compensated by a subsequent increase in its amount. However, the neural mechanisms underlying the homeostatic control of REMs are largely unknown. Here, we show that GABAergic neurons in the preoptic area of the hypothalamus projecting to the tuberomammillary nucleus (POAGAD2→TMN neurons) are crucial for the homeostatic regulation of REMs in mice. POAGAD2→TMN neurons are most active during REMs, and inhibiting them specifically decreases REMs. REMs restriction leads to an increased number and amplitude of calcium transients in POAGAD2→TMN neurons, reflecting the accumulation of REMs pressure. Inhibiting POAGAD2→TMN neurons during REMs restriction blocked the subsequent rebound of REMs. Our findings reveal a hypothalamic circuit whose activity mirrors the buildup of homeostatic REMs pressure during restriction and that is required for the ensuing rebound in REMs.
Collapse
Affiliation(s)
- John J Maurer
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Alexandra Lin
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Xi Jin
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Jiso Hong
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Nicholas Sathi
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Romain Cardis
- Department of Fundamental Neurosciences, University of LausanneLausanneSwitzerland
| | | | - Anita Lüthi
- Department of Fundamental Neurosciences, University of LausanneLausanneSwitzerland
| | - Franz Weber
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Shinjae Chung
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
19
|
Gonzalez Burgos G, Miyamae T, Nishihata Y, Krimer OL, Wade K, Fish KN, Arion D, Cai ZL, Xue M, Stauffer WR, Lewis DA. Synaptic alterations in pyramidal cells following genetic manipulation of neuronal excitability in monkey prefrontal cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598658. [PMID: 38915638 PMCID: PMC11195287 DOI: 10.1101/2024.06.12.598658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
In schizophrenia, layer 3 pyramidal neurons (L3PNs) in the dorsolateral prefrontal cortex (DLPFC) are thought to receive fewer excitatory synaptic inputs and to have lower expression levels of activity-dependent genes and of genes involved in mitochondrial energy production. In concert, these findings from previous studies suggest that DLPFC L3PNs are hypoactive in schizophrenia, disrupting the patterns of activity that are crucial for working memory, which is impaired in the illness. However, whether lower PN activity produces alterations in inhibitory and/or excitatory synaptic strength has not been tested in the primate DLPFC. Here, we decreased PN excitability in rhesus monkey DLPFC in vivo using adeno-associated viral vectors (AAVs) to produce Cre recombinase-mediated overexpression of Kir2.1 channels, a genetic silencing tool that efficiently decreases neuronal excitability. In acute slices prepared from DLPFC 7-12 weeks post-AAV microinjections, Kir2.1-overexpressing PNs had a significantly reduced excitability largely attributable to highly specific effects of the AAV-encoded Kir2.1 channels. Moreover, recordings of synaptic currents showed that Kir2.1-overexpressing DLPFC PNs had reduced strength of excitatory synapses whereas inhibitory synaptic inputs were not affected. The decrease in excitatory synaptic strength was not associated with changes in dendritic spine number, suggesting that excitatory synapse quantity was unaltered in Kir2.1-overexpressing DLPFC PNs. These findings suggest that, in schizophrenia, the excitatory synapses on hypoactive L3PNs are weaker and thus might represent a substrate for novel therapeutic interventions. Significance Statement In schizophrenia, dorsolateral prefrontal cortex (DLPFC) pyramidal neurons (PNs) have both transcriptional and structural alterations that suggest they are hypoactive. PN hypoactivity is thought to produce synaptic alterations in schizophrenia, however the effects of lower neuronal activity on synaptic function in primate DLPFC have not been examined. Here, we used, for the first time in primate neocortex, adeno-associated viral vectors (AAVs) to reduce PN excitability with Kir2.1 channel overexpression and tested if this manipulation altered the strength of synaptic inputs onto the Kir2.1-overexpressing PNs. Recordings in DLPFC slices showed that Kir2.1 overexpression depressed excitatory (but not inhibitory), synaptic currents, suggesting that, in schizophrenia, the hypoactivity of PNs might be exacerbated by reduced strength of the excitatory synapses they receive.
Collapse
|
20
|
Driscoll N, Antonini MJ, Cannon TM, Maretich P, Olaitan G, Phi Van VD, Nagao K, Sahasrabudhe A, Vargas E, Hunt S, Hummel M, Mupparaju S, Jasanoff A, Venton J, Anikeeva P. Fiber-based Probes for Electrophysiology, Photometry, Optical and Electrical Stimulation, Drug Delivery, and Fast-Scan Cyclic Voltammetry In Vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.07.598004. [PMID: 38895451 PMCID: PMC11185794 DOI: 10.1101/2024.06.07.598004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Recording and modulation of neuronal activity enables the study of brain function in health and disease. While translational neuroscience relies on electrical recording and modulation techniques, mechanistic studies in rodent models leverage genetic precision of optical methods, such as optogenetics and imaging of fluorescent indicators. In addition to electrical signal transduction, neurons produce and receive diverse chemical signals which motivate tools to probe and modulate neurochemistry. Although the past decade has delivered a wealth of technologies for electrophysiology, optogenetics, chemical sensing, and optical recording, combining these modalities within a single platform remains challenging. This work leverages materials selection and convergence fiber drawing to permit neural recording, electrical stimulation, optogenetics, fiber photometry, drug and gene delivery, and voltammetric recording of neurotransmitters within individual fibers. Composed of polymers and non-magnetic carbon-based conductors, these fibers are compatible with magnetic resonance imaging, enabling concurrent stimulation and whole-brain monitoring. Their utility is demonstrated in studies of the mesolimbic reward pathway by simultaneously interfacing with the ventral tegmental area and nucleus accumbens in mice and characterizing the neurophysiological effects of a stimulant drug. This study highlights the potential of these fibers to probe electrical, optical, and chemical signaling across multiple brain regions in both mechanistic and translational studies.
Collapse
Affiliation(s)
| | | | | | - Pema Maretich
- Massachusetts Institute of Technology, Cambridge, MA 02139
| | | | | | - Keisuke Nagao
- Massachusetts Institute of Technology, Cambridge, MA 02139
| | | | | | | | - Melissa Hummel
- Massachusetts Institute of Technology, Cambridge, MA 02139
| | | | - Alan Jasanoff
- Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Jill Venton
- The University of Virginia, Charlottesville, VA 22904
| | | |
Collapse
|
21
|
Concetti C, Viskaitis P, Grujic N, Duss SN, Privitera M, Bohacek J, Peleg-Raibstein D, Burdakov D. Exploratory Rearing Is Governed by Hypothalamic Melanin-Concentrating Hormone Neurons According to Locus Ceruleus. J Neurosci 2024; 44:e0015242024. [PMID: 38575343 PMCID: PMC11112542 DOI: 10.1523/jneurosci.0015-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/06/2024] Open
Abstract
Information seeking, such as standing on tiptoes to look around in humans, is observed across animals and helps survival. Its rodent analog-unsupported rearing on hind legs-was a classic model in deciphering neural signals of cognition and is of intense renewed interest in preclinical modeling of neuropsychiatric states. Neural signals and circuits controlling this dedicated decision to seek information remain largely unknown. While studying subsecond timing of spontaneous behavioral acts and activity of melanin-concentrating hormone (MCH) neurons (MNs) in behaving male and female mice, we observed large MN activity spikes that aligned to unsupported rears. Complementary causal, loss and gain of function, analyses revealed specific control of rear frequency and duration by MNs and MCHR1 receptors. Activity in a key stress center of the brain-the locus ceruleus noradrenaline cells-rapidly inhibited MNs and required functional MCH receptors for its endogenous modulation of rearing. By defining a neural module that both tracks and controls rearing, these findings may facilitate further insights into biology of information seeking.
Collapse
Affiliation(s)
- Cristina Concetti
- Department of Health Sciences and Technology, Neuroscience Center Zürich (ZNZ), Swiss Federal Institute of Technology (ETH Zürich), Zürich 8092, Switzerland
| | - Paulius Viskaitis
- Department of Health Sciences and Technology, Neuroscience Center Zürich (ZNZ), Swiss Federal Institute of Technology (ETH Zürich), Zürich 8092, Switzerland
| | - Nikola Grujic
- Department of Health Sciences and Technology, Neuroscience Center Zürich (ZNZ), Swiss Federal Institute of Technology (ETH Zürich), Zürich 8092, Switzerland
| | - Sian N Duss
- Department of Health Sciences and Technology, Neuroscience Center Zürich (ZNZ), Swiss Federal Institute of Technology (ETH Zürich), Zürich 8092, Switzerland
| | - Mattia Privitera
- Department of Health Sciences and Technology, Neuroscience Center Zürich (ZNZ), Swiss Federal Institute of Technology (ETH Zürich), Zürich 8092, Switzerland
| | - Johannes Bohacek
- Department of Health Sciences and Technology, Neuroscience Center Zürich (ZNZ), Swiss Federal Institute of Technology (ETH Zürich), Zürich 8092, Switzerland
| | - Daria Peleg-Raibstein
- Department of Health Sciences and Technology, Neuroscience Center Zürich (ZNZ), Swiss Federal Institute of Technology (ETH Zürich), Zürich 8092, Switzerland
| | - Denis Burdakov
- Department of Health Sciences and Technology, Neuroscience Center Zürich (ZNZ), Swiss Federal Institute of Technology (ETH Zürich), Zürich 8092, Switzerland
| |
Collapse
|
22
|
Burman RJ, Diviney T, Călin A, Gothard G, Jouhanneau JSM, Poulet JFA, Sen A, Akerman CJ. Optogenetic Determination of Dynamic and Cell-Type-Specific Inhibitory Reversal Potentials. J Neurosci 2024; 44:e1392232024. [PMID: 38604778 PMCID: PMC11097265 DOI: 10.1523/jneurosci.1392-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 03/31/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024] Open
Abstract
The reversal potential refers to the membrane potential at which the net current flow through a channel reverses direction. The reversal potential is determined by transmembrane ion gradients and, in turn, determines how the channel's activity will affect the membrane potential. Traditional investigation into the reversal potential of inhibitory ligand-gated ion channels (EInh) has relied upon the activation of endogenous receptors, such as the GABA-A receptor (GABAAR). There are, however, challenges associated with activating endogenous receptors, including agonist delivery, isolating channel responses, and the effects of receptor saturation and desensitization. Here, we demonstrate the utility of using a light-gated anion channel, stGtACR2, to probe EInh in the rodent brain. Using mice of both sexes, we demonstrate that the properties of this optically activated channel make it a suitable proxy for studying GABAAR receptor-mediated inhibition. We validate this agonist-independent optogenetic strategy in vitro and in vivo and further show how it can accurately capture differences in EInh dynamics following manipulations of endogenous ion fluxes. This allows us to explore distinct resting EInh differences across genetically defined neuronal subpopulations. Using this approach to challenge ion homeostasis mechanisms in neurons, we uncover cell-specific EInh dynamics that are supported by the differential expression of endogenous ion handling mechanisms. Our findings therefore establish an effective optical strategy for revealing novel aspects of inhibitory reversal potentials and thereby expand the repertoire of optogenetics.
Collapse
Affiliation(s)
- Richard J Burman
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, United Kingdom
- Oxford Epilepsy Research Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, United Kingdom
| | - Tara Diviney
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, United Kingdom
| | - Alexandru Călin
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, United Kingdom
| | - Gemma Gothard
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, United Kingdom
| | - Jean-Sébastien M Jouhanneau
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 13125, Germany
- Neuroscience Research Center, Charité-Universitätsmedizin, Berlin 10117, Germany
| | - James F A Poulet
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 13125, Germany
- Neuroscience Research Center, Charité-Universitätsmedizin, Berlin 10117, Germany
| | - Arjune Sen
- Oxford Epilepsy Research Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, United Kingdom
| | - Colin J Akerman
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, United Kingdom
| |
Collapse
|
23
|
Romussi S, Giunti S, Andersen N, De Rosa MJ. C. elegans: a prominent platform for modeling and drug screening in neurological disorders. Expert Opin Drug Discov 2024; 19:565-585. [PMID: 38509691 DOI: 10.1080/17460441.2024.2329103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/06/2024] [Indexed: 03/22/2024]
Abstract
INTRODUCTION Human neurodevelopmental and neurodegenerative diseases (NDevDs and NDegDs, respectively) encompass a broad spectrum of disorders affecting the nervous system with an increasing incidence. In this context, the nematode C. elegans, has emerged as a benchmark model for biological research, especially in the field of neuroscience. AREAS COVERED The authors highlight the numerous advantages of this tiny worm as a model for exploring nervous system pathologies and as a platform for drug discovery. There is a particular focus given to describing the existing models of C. elegans for the study of NDevDs and NDegDs. Specifically, the authors underscore their strong applicability in preclinical drug development. Furthermore, they place particular emphasis on detailing the common techniques employed to explore the nervous system in both healthy and diseased states. EXPERT OPINION Drug discovery constitutes a long and expensive process. The incorporation of invertebrate models, such as C. elegans, stands as an exemplary strategy for mitigating costs and expediting timelines. The utilization of C. elegans as a platform to replicate nervous system pathologies and conduct high-throughput automated assays in the initial phases of drug discovery is pivotal for rendering therapeutic options more attainable and cost-effective.
Collapse
Affiliation(s)
- Stefano Romussi
- Laboratorio de Neurobiología de Invertebrados, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), UNS-CONICET, Bahía Blanca, Argentina
| | - Sebastián Giunti
- Laboratorio de Neurobiología de Invertebrados, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), UNS-CONICET, Bahía Blanca, Argentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - Natalia Andersen
- Laboratorio de Neurobiología de Invertebrados, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), UNS-CONICET, Bahía Blanca, Argentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - María José De Rosa
- Laboratorio de Neurobiología de Invertebrados, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), UNS-CONICET, Bahía Blanca, Argentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| |
Collapse
|
24
|
Ott S, Xu S, Lee N, Hong I, Anns J, Suresh DD, Zhang Z, Zhang X, Harion R, Ye W, Chandramouli V, Jesuthasan S, Saheki Y, Claridge-Chang A. Kalium channelrhodopsins effectively inhibit neurons. Nat Commun 2024; 15:3480. [PMID: 38658537 PMCID: PMC11043423 DOI: 10.1038/s41467-024-47203-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 03/18/2024] [Indexed: 04/26/2024] Open
Abstract
The analysis of neural circuits has been revolutionized by optogenetic methods. Light-gated chloride-conducting anion channelrhodopsins (ACRs)-recently emerged as powerful neuron inhibitors. For cells or sub-neuronal compartments with high intracellular chloride concentrations, however, a chloride conductance can have instead an activating effect. The recently discovered light-gated, potassium-conducting, kalium channelrhodopsins (KCRs) might serve as an alternative in these situations, with potentially broad application. As yet, KCRs have not been shown to confer potent inhibitory effects in small genetically tractable animals. Here, we evaluated the utility of KCRs to suppress behavior and inhibit neural activity in Drosophila, Caenorhabditis elegans, and zebrafish. In direct comparisons with ACR1, a KCR1 variant with enhanced plasma-membrane trafficking displayed comparable potency, but with improved properties that include reduced toxicity and superior efficacy in putative high-chloride cells. This comparative analysis of behavioral inhibition between chloride- and potassium-selective silencing tools establishes KCRs as next-generation optogenetic inhibitors for in vivo circuit analysis in behaving animals.
Collapse
Affiliation(s)
- Stanislav Ott
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Sangyu Xu
- Institute for Molecular and Cell Biology, A*STAR Agency for Science, Technology and Research, Singapore, Singapore
| | - Nicole Lee
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Ivan Hong
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Jonathan Anns
- Institute for Molecular and Cell Biology, A*STAR Agency for Science, Technology and Research, Singapore, Singapore
- School of Biological Sciences and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Danesha Devini Suresh
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Zhiyi Zhang
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Xianyuan Zhang
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Raihanah Harion
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Weiying Ye
- Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Vaishnavi Chandramouli
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Suresh Jesuthasan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Yasunori Saheki
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Adam Claridge-Chang
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore.
- Institute for Molecular and Cell Biology, A*STAR Agency for Science, Technology and Research, Singapore, Singapore.
- Department of Physiology, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
25
|
Bredenberg C, Savin C, Kiani R. Recurrent Neural Circuits Overcome Partial Inactivation by Compensation and Re-learning. J Neurosci 2024; 44:e1635232024. [PMID: 38413233 PMCID: PMC11026338 DOI: 10.1523/jneurosci.1635-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/14/2024] [Accepted: 01/20/2024] [Indexed: 02/29/2024] Open
Abstract
Technical advances in artificial manipulation of neural activity have precipitated a surge in studying the causal contribution of brain circuits to cognition and behavior. However, complexities of neural circuits challenge interpretation of experimental results, necessitating new theoretical frameworks for reasoning about causal effects. Here, we take a step in this direction, through the lens of recurrent neural networks trained to perform perceptual decisions. We show that understanding the dynamical system structure that underlies network solutions provides a precise account for the magnitude of behavioral effects due to perturbations. Our framework explains past empirical observations by clarifying the most sensitive features of behavior, and how complex circuits compensate and adapt to perturbations. In the process, we also identify strategies that can improve the interpretability of inactivation experiments.
Collapse
Affiliation(s)
- Colin Bredenberg
- Center for Neural Science, New York University, New York, NY 10003
| | - Cristina Savin
- Center for Neural Science, New York University, New York, NY 10003
- Center for Data Science, New York University, New York, NY 10011
| | - Roozbeh Kiani
- Center for Neural Science, New York University, New York, NY 10003
- Department of Psychology, New York University, New York, NY 10003
| |
Collapse
|
26
|
Clark AM, Ingold A, Reiche CF, Cundy D, Balsor JL, Federer F, McAlinden N, Cheng Y, Rolston JD, Rieth L, Dawson MD, Mathieson K, Blair S, Angelucci A. An optrode array for spatiotemporally-precise large-scale optogenetic stimulation of deep cortical layers in non-human primates. Commun Biol 2024; 7:329. [PMID: 38485764 PMCID: PMC10940688 DOI: 10.1038/s42003-024-05984-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/27/2024] [Indexed: 03/18/2024] Open
Abstract
Optogenetics has transformed studies of neural circuit function, but remains challenging to apply to non-human primates (NHPs). A major challenge is delivering intense, spatiotemporally-precise, patterned photostimulation across large volumes in deep tissue. Such stimulation is critical, for example, to modulate selectively deep-layer corticocortical feedback circuits. To address this need, we have developed the Utah Optrode Array (UOA), a 10×10 glass needle waveguide array fabricated atop a novel opaque optical interposer, and bonded to an electrically addressable µLED array. In vivo experiments with the UOA demonstrated large-scale, spatiotemporally precise, activation of deep circuits in NHP cortex. Specifically, the UOA permitted both focal (confined to single layers/columns), and widespread (multiple layers/columns) optogenetic activation of deep layer neurons, as assessed with multi-channel laminar electrode arrays, simply by varying the number of activated µLEDs and/or the irradiance. Thus, the UOA represents a powerful optoelectronic device for targeted manipulation of deep-layer circuits in NHP models.
Collapse
Affiliation(s)
- Andrew M Clark
- Department of Ophthalmology and Visual Science, Moran Eye Institute, University of Utah, Salt Lake City, UT, USA
| | - Alexander Ingold
- Department of Ophthalmology and Visual Science, Moran Eye Institute, University of Utah, Salt Lake City, UT, USA
| | - Christopher F Reiche
- Department of Electrical and Computer Engineering, University of Utah, Salt Lake City, UT, USA
| | - Donald Cundy
- Department of Ophthalmology and Visual Science, Moran Eye Institute, University of Utah, Salt Lake City, UT, USA
| | - Justin L Balsor
- Department of Ophthalmology and Visual Science, Moran Eye Institute, University of Utah, Salt Lake City, UT, USA
| | - Frederick Federer
- Department of Ophthalmology and Visual Science, Moran Eye Institute, University of Utah, Salt Lake City, UT, USA
| | - Niall McAlinden
- SUPA, Institute of Photonics, Department of Physics, University of Strathclyde, Glasgow, UK
| | - Yunzhou Cheng
- SUPA, Institute of Photonics, Department of Physics, University of Strathclyde, Glasgow, UK
| | - John D Rolston
- Departments of Neurosurgery and Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
- Department of Neurosurgery, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Loren Rieth
- Mechanical and Aerospace Engineering, West Virginia University, Morgantown, WV, USA
- Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Martin D Dawson
- SUPA, Institute of Photonics, Department of Physics, University of Strathclyde, Glasgow, UK
| | - Keith Mathieson
- SUPA, Institute of Photonics, Department of Physics, University of Strathclyde, Glasgow, UK
| | - Steve Blair
- Department of Electrical and Computer Engineering, University of Utah, Salt Lake City, UT, USA.
| | - Alessandra Angelucci
- Department of Ophthalmology and Visual Science, Moran Eye Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
27
|
Nikitin ES, Postnikova TY, Proskurina EY, Borodinova AA, Ivanova V, Roshchin MV, Smirnova MP, Kelmanson I, Belousov VV, Balaban PM, Zaitsev AV. Overexpression of KCNN4 channels in principal neurons produces an anti-seizure effect without reducing their coding ability. Gene Ther 2024; 31:144-153. [PMID: 37968509 DOI: 10.1038/s41434-023-00427-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 10/13/2023] [Accepted: 10/24/2023] [Indexed: 11/17/2023]
Abstract
Gene therapy offers a potential alternative to the surgical treatment of epilepsy, which affects millions of people and is pharmacoresistant in ~30% of cases. Aimed at reducing the excitability of principal neurons, the engineered expression of K+ channels has been proposed as a treatment due to the outstanding ability of K+ channels to hyperpolarize neurons. However, the effects of K+ channel overexpression on cell physiology remain to be investigated. Here we report an adeno-associated virus (AAV) vector designed to reduce epileptiform activity specifically in excitatory pyramidal neurons by expressing the human Ca2+-gated K+ channel KCNN4 (KCa3.1). Electrophysiological and pharmacological experiments in acute brain slices showed that KCNN4-transduced cells exhibited a Ca2+-dependent slow afterhyperpolarization that significantly decreased the ability of KCNN4-positive neurons to generate high-frequency spike trains without affecting their lower-frequency coding ability and action potential shapes. Antiepileptic activity tests showed potent suppression of pharmacologically induced seizures in vitro at both single cell and local field potential levels with decreased spiking during ictal discharges. Taken together, our findings strongly suggest that the AAV-based expression of the KCNN4 channel in excitatory neurons is a promising therapeutic intervention as gene therapy for epilepsy.
Collapse
Affiliation(s)
- Evgeny S Nikitin
- Institute of Higher Nervous Activity and Neurophysiology, RAS, 117485, Moscow, Russia.
| | - Tatiana Y Postnikova
- Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, 194223, Saint Petersburg, Russia
| | - Elena Y Proskurina
- Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, 194223, Saint Petersburg, Russia
| | | | - Violetta Ivanova
- Institute of Higher Nervous Activity and Neurophysiology, RAS, 117485, Moscow, Russia
| | - Matvey V Roshchin
- Institute of Higher Nervous Activity and Neurophysiology, RAS, 117485, Moscow, Russia
| | - Maria P Smirnova
- Institute of Higher Nervous Activity and Neurophysiology, RAS, 117485, Moscow, Russia
| | - Ilya Kelmanson
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997, Moscow, Russia
| | - Vsevolod V Belousov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117997, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, 143025, Moscow, Russia
| | - Pavel M Balaban
- Institute of Higher Nervous Activity and Neurophysiology, RAS, 117485, Moscow, Russia
| | - Aleksey V Zaitsev
- Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, 194223, Saint Petersburg, Russia.
| |
Collapse
|
28
|
Agoitia A, Cruz-Sanchez A, Balderas I, Bermúdez-Rattoni F. The anterior insula and its projection to amygdala nuclei modulate the abstinence-exacerbated expression of conditioned place preference. Psychopharmacology (Berl) 2024; 241:445-459. [PMID: 38010515 PMCID: PMC10884150 DOI: 10.1007/s00213-023-06499-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/05/2023] [Indexed: 11/29/2023]
Abstract
RATIONALE Relapse into substance use is often triggered by exposure to drug-related environmental cues. The magnitude of drug seeking depends on the duration of abstinence, a phenomenon known as the incubation of drug craving. Clinical and preclinical research shows that the insular cortex is involved in substance use disorders and cue-induced drug seeking. However, the role of the insula on memory retrieval and motivational integration for cue-elicited drug seeking remains to be determined. OBJECTIVES We investigated the role of the anterior insular cortex (aIC) and its glutamatergic projection to amygdala nuclei (aIC-AMY) on the expression of conditioned place preference (CPP) during early and late abstinence. METHODS Male adult C57BL/6J mice underwent amphetamine-induced CPP, and their preference was tested following 1 or 14 days of abstinence. aIC and aIC-AMY functional role in CPP expression was assessed at both abstinence periods by employing optogenetic silencing and behavioral pharmacology. RESULTS Compared to a single day, an exacerbated preference for the amphetamine-paired context was observed after 14 days of abstinence. Photoinhibition of either aIC or aIC-AMY projection reduced CPP expression following late but not early abstinence. Similarly, the antagonism of aIC NMDA receptors reduced CPP expression after 14 days of abstinence but not 1 day. CONCLUSIONS These results suggest that aIC and its glutamatergic output to amygdala nuclei constitute critical neurobiological substrates mediating enhanced motivational cue reactivity during the incubation of amphetamine craving rather than contextual memory recall. Moreover, cortical NMDA receptor signaling may become sensitized during abstinence, ultimately modulating disproportioned drug seeking.
Collapse
Affiliation(s)
- Andrés Agoitia
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Apolinar Cruz-Sanchez
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Israela Balderas
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Federico Bermúdez-Rattoni
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.
| |
Collapse
|
29
|
Palchaudhuri S, Osypenko D, Schneggenburger R. Fear Learning: An Evolving Picture for Plasticity at Synaptic Afferents to the Amygdala. Neuroscientist 2024; 30:87-104. [PMID: 35822657 DOI: 10.1177/10738584221108083] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Unraveling the neuronal mechanisms of fear learning might allow neuroscientists to make links between a learned behavior and the underlying plasticity at specific synaptic connections. In fear learning, an innocuous sensory event such as a tone (called the conditioned stimulus, CS) acquires an emotional value when paired with an aversive outcome (unconditioned stimulus, US). Here, we review earlier studies that have shown that synaptic plasticity at thalamic and cortical afferents to the lateral amygdala (LA) is critical for the formation of auditory-cued fear memories. Despite the early progress, it has remained unclear whether there are separate synaptic inputs that carry US information to the LA to act as a teaching signal for plasticity at CS-coding synapses. Recent findings have begun to fill this gap by showing, first, that thalamic and cortical auditory afferents can also carry US information; second, that the release of neuromodulators contributes to US-driven teaching signals; and third, that synaptic plasticity additionally happens at connections up- and downstream of the LA. Together, a picture emerges in which coordinated synaptic plasticity in serial and parallel circuits enables the formation of a finely regulated fear memory.
Collapse
Affiliation(s)
- Shriya Palchaudhuri
- Laboratory of Synaptic Mechanisms, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Denys Osypenko
- Laboratory of Synaptic Mechanisms, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Ralf Schneggenburger
- Laboratory of Synaptic Mechanisms, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
30
|
Li D, Yan X, Xing Y, Yan J, Wang J, Zhang H, Wang J, Li X, Su Z, Loh HH, Yang X, Chen X. Local and Remote Chemogenetic Suppression of Hippocampal Seizures in Rats. Curr Neuropharmacol 2024; 22:2240-2255. [PMID: 38333970 PMCID: PMC11337692 DOI: 10.2174/1570159x22999240131122455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 05/24/2023] [Accepted: 06/06/2023] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Innovative treatments of refractory epilepsy are widely desired, for which chemogenetic technology can provide region- and cell-type-specific modulation with relative noninvasiveness. OBJECTIVES We aimed to explore the specific applications of chemogenetics for locally and remotely networks controlling hippocampal seizures. METHODS A virus coding for a modified human Gi-coupled M4 muscarinic receptor (hM4Di) on pyramidal cells was injected into either the right hippocampal CA3 or the bilateral anterior nucleus of the thalamus (ANT) in rats. After one month, seizures were induced by 4-aminopyridine (4-AP) injection into the right CA3. Simultaneously, clozapine-N-oxide (CNO) (2.5 mg/kg) or clozapine (0.1 mg/kg), the specific ligands acting on hM4Di, were injected intraperitoneally. We also set up hM4Di control and clozapine control groups to eliminate the influence of viral transfection and the ligand alone on the experimental results. RESULTS For both local and remote controls, the mean seizure duration was significantly reduced upon ligand application in the experimental groups. Seizure frequency, on the other hand, only showed a significant decrease in local control, with a lower frequency in the clozapine group than in the CNO group. Both the effects of CNO and clozapine were time-dependent, and clozapine was faster than CNO in local seizure control. CONCLUSION This study shows the potency of chemogenetics to attenuate hippocampal seizures locally or remotely by activating the transfected hM4Di receptor with CNO or clozapine. ANT is suggested as a potentially safe chemogenetic application target in the epileptic network for focal hippocampal seizures.
Collapse
Affiliation(s)
- Donghong Li
- Department of Neurology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xi Yan
- Department of Neurology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, Henan, China
| | - Yue Xing
- Guangzhou Laboratory, Guangzhou, Guangdong, China
| | - Jiaqing Yan
- College of Electrical and Control Engineering, North China University of Technology, Beijing, China
| | - Junling Wang
- Guangzhou Laboratory, Guangzhou, Guangdong, China
| | - Herui Zhang
- Guangzhou Laboratory, Guangzhou, Guangdong, China
| | | | - Xiaonan Li
- Guangzhou Laboratory, Guangzhou, Guangdong, China
| | - Zhumin Su
- Department of Neurology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | | | | | - Xiaohong Chen
- Department of Neurology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
31
|
Zhou F, Tichy AM, Imambocus BN, Sakharwade S, Rodriguez Jimenez FJ, González Martínez M, Jahan I, Habib M, Wilhelmy N, Burre V, Lömker T, Sauter K, Helfrich-Förster C, Pielage J, Grunwald Kadow IC, Janovjak H, Soba P. Optimized design and in vivo application of optogenetically functionalized Drosophila dopamine receptors. Nat Commun 2023; 14:8434. [PMID: 38114457 PMCID: PMC10730509 DOI: 10.1038/s41467-023-43970-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 11/24/2023] [Indexed: 12/21/2023] Open
Abstract
Neuromodulatory signaling via G protein-coupled receptors (GPCRs) plays a pivotal role in regulating neural network function and animal behavior. The recent development of optogenetic tools to induce G protein-mediated signaling provides the promise of acute and cell type-specific manipulation of neuromodulatory signals. However, designing and deploying optogenetically functionalized GPCRs (optoXRs) with accurate specificity and activity to mimic endogenous signaling in vivo remains challenging. Here we optimize the design of optoXRs by considering evolutionary conserved GPCR-G protein interactions and demonstrate the feasibility of this approach using two Drosophila Dopamine receptors (optoDopRs). These optoDopRs exhibit high signaling specificity and light sensitivity in vitro. In vivo, we show receptor and cell type-specific effects of dopaminergic signaling in various behaviors, including the ability of optoDopRs to rescue the loss of the endogenous receptors. This work demonstrates that optoXRs can enable optical control of neuromodulatory receptor-specific signaling in functional and behavioral studies.
Collapse
Affiliation(s)
- Fangmin Zhou
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115, Bonn, Germany
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Alexandra-Madelaine Tichy
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, 3800, Clayton, Victoria, Australia
- European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, 3800, Clayton, Victoria, Australia
| | - Bibi Nusreen Imambocus
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115, Bonn, Germany
| | - Shreyas Sakharwade
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115, Bonn, Germany
| | - Francisco J Rodriguez Jimenez
- Institute of Physiology II, University Clinic Bonn (UKB), University of Bonn, 53115, Bonn, Germany
- ZIEL-Institute of Life and Health, Technical University of Munich, School of Life Sciences, 85354, Freising, Germany
| | - Marco González Martínez
- Institute of Physiology II, University Clinic Bonn (UKB), University of Bonn, 53115, Bonn, Germany
| | - Ishrat Jahan
- Institute of Physiology II, University Clinic Bonn (UKB), University of Bonn, 53115, Bonn, Germany
| | - Margarita Habib
- Neurobiology and Genetics, Biocenter, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Nina Wilhelmy
- Division of Neurobiology and Zoology, RPTU University of Kaiserslautern, 67663, Kaiserslautern, Germany
| | - Vanessa Burre
- Division of Neurobiology and Zoology, RPTU University of Kaiserslautern, 67663, Kaiserslautern, Germany
| | - Tatjana Lömker
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Kathrin Sauter
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | | | - Jan Pielage
- Division of Neurobiology and Zoology, RPTU University of Kaiserslautern, 67663, Kaiserslautern, Germany
| | - Ilona C Grunwald Kadow
- Institute of Physiology II, University Clinic Bonn (UKB), University of Bonn, 53115, Bonn, Germany
- ZIEL-Institute of Life and Health, Technical University of Munich, School of Life Sciences, 85354, Freising, Germany
| | - Harald Janovjak
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, 3800, Clayton, Victoria, Australia
- European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, 3800, Clayton, Victoria, Australia
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, 5042, Bedford Park, South Australia, Australia
| | - Peter Soba
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany.
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115, Bonn, Germany.
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
| |
Collapse
|
32
|
Hoffmann C, Milovanovic D. Dipping contacts - a novel type of contact site at the interface between membraneless organelles and membranes. J Cell Sci 2023; 136:jcs261413. [PMID: 38149872 PMCID: PMC10785658 DOI: 10.1242/jcs.261413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023] Open
Abstract
Liquid-liquid phase separation is a major mechanism for organizing macromolecules, particularly proteins with intrinsically disordered regions, in compartments not limited by a membrane or a scaffold. The cell can therefore be perceived as a complex emulsion containing many of these membraneless organelles, also referred to as biomolecular condensates, together with numerous membrane-bound organelles. It is currently unclear how such a complex concoction operates to allow for intracellular trafficking, signaling and metabolic processes to occur with high spatiotemporal precision. Based on experimental observations of synaptic vesicle condensates - a membraneless organelle that is in fact packed with membranes - we present here the framework of dipping contacts: a novel type of contact site between membraneless organelles and membranes. In this Hypothesis, we propose that our framework of dipping contacts can serve as a foundation to investigate the interface that couples the diffusion and material properties of condensates to biochemical processes occurring in membranes. The identity and regulation of this interface is especially critical in the case of neurodegenerative diseases, where aberrant inclusions of misfolded proteins and damaged organelles underlie cellular pathology.
Collapse
Affiliation(s)
- Christian Hoffmann
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Whitman Center, Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Dragomir Milovanovic
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Whitman Center, Marine Biological Laboratory, Woods Hole, MA 02543, USA
- National Center for X-ray Tomography, Advanced Light Source, Berkeley, CA 94720, USA
- Einstein Center for Neuroscience, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität Berlin and Berlin Institute of Health, 10117 Berlin, Germany
| |
Collapse
|
33
|
Mirabella PN, Fenselau H. Advanced neurobiological tools to interrogate metabolism. Nat Rev Endocrinol 2023; 19:639-654. [PMID: 37674015 DOI: 10.1038/s41574-023-00885-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/24/2023] [Indexed: 09/08/2023]
Abstract
Engineered neurobiological tools for the manipulation of cellular activity, such as chemogenetics and optogenetics, have become a cornerstone of modern neuroscience research. These tools are invaluable for the interrogation of the central control of metabolism as they provide a direct means to establish a causal relationship between brain activity and biological processes at the cellular, tissue and organismal levels. The utility of these methods has grown substantially due to advances in cellular-targeting strategies, alongside improvements in the resolution and potency of such tools. Furthermore, the potential to recapitulate endogenous cellular signalling has been enriched by insights into the molecular signatures and activity dynamics of discrete brain cell types. However, each modulatory tool has a specific set of advantages and limitations; therefore, tool selection and suitability are of paramount importance to optimally interrogate the cellular and circuit-based underpinnings of metabolic outcomes within the organism. Here, we describe the key principles and uses of engineered neurobiological tools. We also highlight inspiring applications and outline critical considerations to be made when using these tools within the field of metabolism research. We contend that the appropriate application of these biotechnological advances will enable the delineation of the central circuitry regulating systemic metabolism with unprecedented potential.
Collapse
Affiliation(s)
- Paul Nicholas Mirabella
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Cologne, Germany
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany
| | - Henning Fenselau
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Cologne, Germany.
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany.
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Cologne, Germany.
| |
Collapse
|
34
|
Marcus DJ, Bruchas MR. Optical Approaches for Investigating Neuromodulation and G Protein-Coupled Receptor Signaling. Pharmacol Rev 2023; 75:1119-1139. [PMID: 37429736 PMCID: PMC10595021 DOI: 10.1124/pharmrev.122.000584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 04/06/2023] [Accepted: 05/01/2023] [Indexed: 07/12/2023] Open
Abstract
Despite the fact that roughly 40% of all US Food and Drug Administration (FDA)-approved pharmacological therapeutics target G protein-coupled receptors (GPCRs), there remains a gap in our understanding of the physiologic and functional role of these receptors at the systems level. Although heterologous expression systems and in vitro assays have revealed a tremendous amount about GPCR signaling cascades, how these cascades interact across cell types, tissues, and organ systems remains obscure. Classic behavioral pharmacology experiments lack both the temporal and spatial resolution to resolve these long-standing issues. Over the past half century, there has been a concerted effort toward the development of optical tools for understanding GPCR signaling. From initial ligand uncaging approaches to more recent development of optogenetic techniques, these strategies have allowed researchers to probe longstanding questions in GPCR pharmacology both in vivo and in vitro. These tools have been employed across biologic systems and have allowed for interrogation of everything from specific intramolecular events to pharmacology at the systems level in a spatiotemporally specific manner. In this review, we present a historical perspective on the motivation behind and development of a variety of optical toolkits that have been generated to probe GPCR signaling. Here we highlight how these tools have been used in vivo to uncover the functional role of distinct populations of GPCRs and their signaling cascades at a systems level. SIGNIFICANCE STATEMENT: G protein-coupled receptors (GPCRs) remain one of the most targeted classes of proteins for pharmaceutical intervention, yet we still have a limited understanding of how their unique signaling cascades effect physiology and behavior at the systems level. In this review, we discuss a vast array of optical techniques that have been devised to probe GPCR signaling both in vitro and in vivo.
Collapse
Affiliation(s)
- David J Marcus
- Center for the Neurobiology of Addiction, Pain and Emotion (D.J.M., M.R.B.), Department of Anesthesiology and Pain Medicine (D.J.M., M.R.B.), Department of Pharmacology (M.R.B.), and Department of Bioengineering (M.R.B.), University of Washington, Seattle, Washington
| | - Michael R Bruchas
- Center for the Neurobiology of Addiction, Pain and Emotion (D.J.M., M.R.B.), Department of Anesthesiology and Pain Medicine (D.J.M., M.R.B.), Department of Pharmacology (M.R.B.), and Department of Bioengineering (M.R.B.), University of Washington, Seattle, Washington
| |
Collapse
|
35
|
Xu S, Momin M, Ahmed S, Hossain A, Veeramuthu L, Pandiyan A, Kuo CC, Zhou T. Illuminating the Brain: Advances and Perspectives in Optoelectronics for Neural Activity Monitoring and Modulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2303267. [PMID: 37726261 DOI: 10.1002/adma.202303267] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/30/2023] [Indexed: 09/21/2023]
Abstract
Optogenetic modulation of brain neural activity that combines optical and electrical modes in a unitary neural system has recently gained robust momentum. Controlling illumination spatial coverage, designing light-activated modulators, and developing wireless light delivery and data transmission are crucial for maximizing the use of optical neuromodulation. To this end, biocompatible electrodes with enhanced optoelectrical performance, device integration for multiplexed addressing, wireless transmission, and multimodal operation in soft systems have been developed. This review provides an outlook for uniformly illuminating large brain areas while spatiotemporally imaging the neural responses upon optoelectrical stimulation with little artifacts. Representative concepts and important breakthroughs, such as head-mounted illumination, multiple implanted optical fibers, and micro-light-delivery devices, are discussed. Examples of techniques that incorporate electrophysiological monitoring and optoelectrical stimulation are presented. Challenges and perspectives are posed for further research efforts toward high-density optoelectrical neural interface modulation, with the potential for nonpharmacological neurological disease treatments and wireless optoelectrical stimulation.
Collapse
Affiliation(s)
- Shumao Xu
- Department of Engineering Science and Mechanics, Center for Neural Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| | - Marzia Momin
- Department of Engineering Science and Mechanics, Center for Neural Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| | - Salahuddin Ahmed
- Department of Engineering Science and Mechanics, Center for Neural Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| | - Arafat Hossain
- Department of Electrical Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| | - Loganathan Veeramuthu
- Department of Molecular Science and Engineering, National Taipei University of Technology, Taipei, 10608, Republic of China
| | - Archana Pandiyan
- Department of Molecular Science and Engineering, National Taipei University of Technology, Taipei, 10608, Republic of China
| | - Chi-Ching Kuo
- Department of Molecular Science and Engineering, National Taipei University of Technology, Taipei, 10608, Republic of China
| | - Tao Zhou
- Department of Engineering Science and Mechanics, Center for Neural Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| |
Collapse
|
36
|
Tajima S, Kim YS, Fukuda M, Jo Y, Wang PY, Paggi JM, Inoue M, Byrne EFX, Kishi KE, Nakamura S, Ramakrishnan C, Takaramoto S, Nagata T, Konno M, Sugiura M, Katayama K, Matsui TE, Yamashita K, Kim S, Ikeda H, Kim J, Kandori H, Dror RO, Inoue K, Deisseroth K, Kato HE. Structural basis for ion selectivity in potassium-selective channelrhodopsins. Cell 2023; 186:4325-4344.e26. [PMID: 37652010 PMCID: PMC7615185 DOI: 10.1016/j.cell.2023.08.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 05/11/2023] [Accepted: 08/07/2023] [Indexed: 09/02/2023]
Abstract
KCR channelrhodopsins (K+-selective light-gated ion channels) have received attention as potential inhibitory optogenetic tools but more broadly pose a fundamental mystery regarding how their K+ selectivity is achieved. Here, we present 2.5-2.7 Å cryo-electron microscopy structures of HcKCR1 and HcKCR2 and of a structure-guided mutant with enhanced K+ selectivity. Structural, electrophysiological, computational, spectroscopic, and biochemical analyses reveal a distinctive mechanism for K+ selectivity; rather than forming the symmetrical filter of canonical K+ channels achieving both selectivity and dehydration, instead, three extracellular-vestibule residues within each monomer form a flexible asymmetric selectivity gate, while a distinct dehydration pathway extends intracellularly. Structural comparisons reveal a retinal-binding pocket that induces retinal rotation (accounting for HcKCR1/HcKCR2 spectral differences), and design of corresponding KCR variants with increased K+ selectivity (KALI-1/KALI-2) provides key advantages for optogenetic inhibition in vitro and in vivo. Thus, discovery of a mechanism for ion-channel K+ selectivity also provides a framework for next-generation optogenetics.
Collapse
Affiliation(s)
- Seiya Tajima
- Komaba Institute for Science, The University of Tokyo, Meguro, Tokyo, Japan
| | - Yoon Seok Kim
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Masahiro Fukuda
- Komaba Institute for Science, The University of Tokyo, Meguro, Tokyo, Japan
| | - YoungJu Jo
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Peter Y Wang
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Joseph M Paggi
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Masatoshi Inoue
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Eamon F X Byrne
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Koichiro E Kishi
- Komaba Institute for Science, The University of Tokyo, Meguro, Tokyo, Japan
| | - Seiwa Nakamura
- Komaba Institute for Science, The University of Tokyo, Meguro, Tokyo, Japan
| | | | - Shunki Takaramoto
- The Institute for Solid State Physics, The University of Tokyo, Kashiwa, Japan
| | - Takashi Nagata
- The Institute for Solid State Physics, The University of Tokyo, Kashiwa, Japan
| | - Masae Konno
- The Institute for Solid State Physics, The University of Tokyo, Kashiwa, Japan; PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
| | - Masahiro Sugiura
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Showa-ku, Japan
| | - Kota Katayama
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Showa-ku, Japan
| | - Toshiki E Matsui
- Komaba Institute for Science, The University of Tokyo, Meguro, Tokyo, Japan
| | - Keitaro Yamashita
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Suhyang Kim
- Komaba Institute for Science, The University of Tokyo, Meguro, Tokyo, Japan
| | - Hisako Ikeda
- Komaba Institute for Science, The University of Tokyo, Meguro, Tokyo, Japan
| | - Jaeah Kim
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Hideki Kandori
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Showa-ku, Japan; OptoBioTechnology Research Center, Nagoya Institute of Technology, Showa-ku, Japan
| | - Ron O Dror
- Department of Computer Science, Stanford University, Stanford, CA, USA; Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Keiichi Inoue
- The Institute for Solid State Physics, The University of Tokyo, Kashiwa, Japan
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA, USA; CNC Program, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA; Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
| | - Hideaki E Kato
- Komaba Institute for Science, The University of Tokyo, Meguro, Tokyo, Japan; Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, Japan; FOREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan.
| |
Collapse
|
37
|
Li Q, Takeuchi Y, Wang J, Gellért L, Barcsai L, Pedraza LK, Nagy AJ, Kozák G, Nakai S, Kato S, Kobayashi K, Ohsawa M, Horváth G, Kékesi G, Lőrincz ML, Devinsky O, Buzsáki G, Berényi A. Reinstating olfactory bulb-derived limbic gamma oscillations alleviates depression-like behavioral deficits in rodents. Neuron 2023; 111:2065-2075.e5. [PMID: 37164008 PMCID: PMC10321244 DOI: 10.1016/j.neuron.2023.04.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 01/31/2023] [Accepted: 04/12/2023] [Indexed: 05/12/2023]
Abstract
Although the etiology of major depressive disorder remains poorly understood, reduced gamma oscillations is an emerging biomarker. Olfactory bulbectomy, an established model of depression that reduces limbic gamma oscillations, suffers from non-specific effects of structural damage. Here, we show that transient functional suppression of olfactory bulb neurons or their piriform cortex efferents decreased gamma oscillation power in limbic areas and induced depression-like behaviors in rodents. Enhancing transmission of gamma oscillations from olfactory bulb to limbic structures by closed-loop electrical neuromodulation alleviated these behaviors. By contrast, silencing gamma transmission by anti-phase closed-loop stimulation strengthened depression-like behaviors in naive animals. These induced behaviors were neutralized by ketamine treatment that restored limbic gamma power. Taken together, our results reveal a causal link between limbic gamma oscillations and depression-like behaviors in rodents. Interfering with these endogenous rhythms can affect behaviors in rodent models of depression, suggesting that restoring gamma oscillations may alleviate depressive symptoms.
Collapse
Affiliation(s)
- Qun Li
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary; HCEMM-SZTE Magnetotherapeutics Research Group, University of Szeged, Szeged 6720, Hungary
| | - Yuichi Takeuchi
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary; Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; Department of Physiology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan; Department of Biopharmaceutical Sciences and Pharmacy, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Jiale Wang
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary; Faculty of Agriculture, University of Szeged, Szeged 6720, Hungary
| | - Levente Gellért
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary; HCEMM-SZTE Magnetotherapeutics Research Group, University of Szeged, Szeged 6720, Hungary
| | - Livia Barcsai
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary; HCEMM-SZTE Magnetotherapeutics Research Group, University of Szeged, Szeged 6720, Hungary; Neunos Inc, Boston, MA 02108, USA
| | - Lizeth K Pedraza
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary
| | - Anett J Nagy
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary; HCEMM-SZTE Magnetotherapeutics Research Group, University of Szeged, Szeged 6720, Hungary; Neunos Inc, Boston, MA 02108, USA
| | - Gábor Kozák
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary
| | - Shinya Nakai
- Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Masahiro Ohsawa
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary; Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Gyöngyi Horváth
- Department of Physiology, University of Szeged, Szeged 6720, Hungary
| | - Gabriella Kékesi
- Department of Physiology, University of Szeged, Szeged 6720, Hungary
| | - Magor L Lőrincz
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary; Department of Physiology, Anatomy and Neuroscience, Faculty of Sciences University of Szeged, Szeged 6726, Hungary; Neuroscience Division, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | - Orrin Devinsky
- Department of Neurology, NYU Langone Comprehensive Epilepsy Center, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - György Buzsáki
- Neuroscience Institute, New York University, New York, NY 10016, USA
| | - Antal Berényi
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary; HCEMM-SZTE Magnetotherapeutics Research Group, University of Szeged, Szeged 6720, Hungary; Neunos Inc, Boston, MA 02108, USA; Neuroscience Institute, New York University, New York, NY 10016, USA.
| |
Collapse
|
38
|
Wietek J, Nozownik A, Pulin M, Saraf-Sinik I, Matosevich N, Malan D, Brown BJ, Dine J, Levy R, Litvin A, Regev N, Subramaniam S, Bitton E, Benjamin A, Copits BA, Sasse P, Rost BR, Schmitz D, Soba P, Nir Y, Wiegert JS, Yizhar O. A bistable inhibitory OptoGPCR for multiplexed optogenetic control of neural circuits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.01.547328. [PMID: 37425961 PMCID: PMC10327178 DOI: 10.1101/2023.07.01.547328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Information is transmitted between brain regions through the release of neurotransmitters from long-range projecting axons. Understanding how the activity of such long-range connections contributes to behavior requires efficient methods for reversibly manipulating their function. Chemogenetic and optogenetic tools, acting through endogenous G-protein coupled receptor (GPCRs) pathways, can be used to modulate synaptic transmission, but existing tools are limited in sensitivity, spatiotemporal precision, or spectral multiplexing capabilities. Here we systematically evaluated multiple bistable opsins for optogenetic applications and found that the Platynereis dumerilii ciliary opsin (PdCO) is an efficient, versatile, light-activated bistable GPCR that can suppress synaptic transmission in mammalian neurons with high temporal precision in-vivo. PdCO has superior biophysical properties that enable spectral multiplexing with other optogenetic actuators and reporters. We demonstrate that PdCO can be used to conduct reversible loss-of-function experiments in long-range projections of behaving animals, thereby enabling detailed synapse-specific functional circuit mapping.
Collapse
Affiliation(s)
- Jonas Wietek
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Adrianna Nozownik
- Center for Molecular Neurobiology, Hamburg, Germany
- Present address: Paris Brain Institute, Institut du Cerveau (ICM), CNRS UMR 7225, INSERM U1127, Sorbonne Université, Paris, France
| | - Mauro Pulin
- Center for Molecular Neurobiology, Hamburg, Germany
- Present address: Laboratory of Sensory Processing, Brain Mind Institute, Faculty of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Inbar Saraf-Sinik
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Noa Matosevich
- Sagol school of neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Daniela Malan
- Institut für Physiologie I, Universität Bonn, Bonn, Germany
| | - Bobbie J. Brown
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Julien Dine
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
- Present address: Boehringer Ingelheim Pharma GmbH & Co. KG; CNS Diseases, Biberach an der Riss, Germany
| | - Rivka Levy
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Anna Litvin
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Noa Regev
- Department of Physiology and Pharmacology, Tel Aviv University, Tel Aviv, Israel
| | - Suraj Subramaniam
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal Bitton
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Asaf Benjamin
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Bryan A. Copits
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Philipp Sasse
- Institut für Physiologie I, Universität Bonn, Bonn, Germany
| | - Benjamin R. Rost
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Neuroscience Research Center, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Dietmar Schmitz
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Neuroscience Research Center, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
- Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Peter Soba
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- LIMES-Institute, University of Bonn, Bonn, Germany
| | - Yuval Nir
- Sagol school of neuroscience, Tel Aviv University, Tel Aviv, Israel
- Department of Physiology and Pharmacology, Tel Aviv University, Tel Aviv, Israel
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - J. Simon Wiegert
- Center for Molecular Neurobiology, Hamburg, Germany
- Present address: MCTN, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Ofer Yizhar
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
39
|
Kawano T, Zhou J, Anwar S, Salah H, Dayal AH, Ishikawa Y, Boetel K, Takahashi T, Sharma K, Inoue M. T cell infiltration into the brain triggers pulmonary dysfunction in murine Cryptococcus-associated IRIS. Nat Commun 2023; 14:3831. [PMID: 37380639 DOI: 10.1038/s41467-023-39518-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 06/16/2023] [Indexed: 06/30/2023] Open
Abstract
Cryptococcus-associated immune reconstitution inflammatory syndrome (C-IRIS) is a condition frequently occurring in immunocompromised patients receiving antiretroviral therapy. C-IRIS patients exhibit many critical symptoms, including pulmonary distress, potentially complicating the progression and recovery from this condition. Here, utilizing our previously established mouse model of unmasking C-IRIS (CnH99 preinfection and adoptive transfer of CD4+ T cells), we demonstrated that pulmonary dysfunction associated with the C-IRIS condition in mice could be attributed to the infiltration of CD4+ T cells into the brain via the CCL8-CCR5 axis, which triggers the nucleus tractus solitarius (NTS) neuronal damage and neuronal disconnection via upregulated ephrin B3 and semaphorin 6B in CD4+ T cells. Our findings provide unique insight into the mechanism behind pulmonary dysfunction in C-IRIS and nominate potential therapeutic targets for treatment.
Collapse
Affiliation(s)
- Tasuku Kawano
- Department of Comparative Biosciences, The University of Illinois at Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima Aoba-Ku, Sendai, Miyagi, 981-8558, Japan
| | - Jinyan Zhou
- Department of Comparative Biosciences, The University of Illinois at Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
- Neuroscience Program, The University of Illinois at Urbana-Champaign, 405 North Matthews Avenue, Urbana, IL, 61801, USA
| | - Shehata Anwar
- Department of Comparative Biosciences, The University of Illinois at Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
- Department of Pathology, Faculty of Veterinary Medicine, Beni-Suef University (BSU), Beni-Suef, 62511, Egypt
| | - Haneen Salah
- Department of Comparative Biosciences, The University of Illinois at Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
- School of Molecular and Cell Biology, The University of Illinois at Urbana-Champaign, 407 South Goodwin Avenue, Urbana, IL, 61801, USA
| | - Andrea H Dayal
- Department of Comparative Biosciences, The University of Illinois at Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
- School of Molecular and Cell Biology, The University of Illinois at Urbana-Champaign, 407 South Goodwin Avenue, Urbana, IL, 61801, USA
| | - Yuzuki Ishikawa
- Department of Comparative Biosciences, The University of Illinois at Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
- School of Molecular and Cell Biology, The University of Illinois at Urbana-Champaign, 407 South Goodwin Avenue, Urbana, IL, 61801, USA
| | - Katelyn Boetel
- Department of Comparative Biosciences, The University of Illinois at Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
- School of Molecular and Cell Biology, The University of Illinois at Urbana-Champaign, 407 South Goodwin Avenue, Urbana, IL, 61801, USA
| | - Tomoko Takahashi
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima Aoba-Ku, Sendai, Miyagi, 981-8558, Japan
| | - Kamal Sharma
- Department of Anatomy and Cell Biology, University of Illinois, Chicago, 808 S. Wood Street, Chicago, IL, 60612, USA
| | - Makoto Inoue
- Department of Comparative Biosciences, The University of Illinois at Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA.
- Neuroscience Program, The University of Illinois at Urbana-Champaign, 405 North Matthews Avenue, Urbana, IL, 61801, USA.
- Beckman Institute for Advanced Science and Technology, 405 North Matthews Avenue, Urbana, IL, 61801, USA.
| |
Collapse
|
40
|
Tsukamoto H, Kubo Y. A self-inactivating invertebrate opsin optically drives biased signaling toward Gβγ-dependent ion channel modulation. Proc Natl Acad Sci U S A 2023; 120:e2301269120. [PMID: 37186850 PMCID: PMC10214182 DOI: 10.1073/pnas.2301269120] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
Animal opsins, light-sensitive G protein-coupled receptors, have been used for optogenetic tools to control G protein-dependent signaling pathways. Upon G protein activation, the Gα and Gβγ subunits drive different intracellular signaling pathways, leading to complex cellular responses. For some purposes, Gα- and Gβγ-dependent signaling needs to be separately modulated, but these responses are simultaneously evoked due to the 1:1 stoichiometry of Gα and Gβγ Nevertheless, we show temporal activation of G protein using a self-inactivating invertebrate opsin, Platynereis c-opsin1, drives biased signaling for Gβγ-dependent GIRK channel activation in a light-dependent manner by utilizing the kinetic difference between Gβγ-dependent and Gα-dependent responses. The opsin-induced transient Gi/o activation preferentially causes activation of the kinetically fast Gβγ-dependent GIRK channels rather than slower Gi/oα-dependent adenylyl cyclase inhibition. Although similar Gβγ-biased signaling properties were observed in a self-inactivating vertebrate visual pigment, Platynereis c-opsin1 requires fewer retinal molecules to evoke cellular responses. Furthermore, the Gβγ-biased signaling properties of Platynereis c-opsin1 are enhanced by genetically fusing with RGS8 protein, which accelerates G protein inactivation. The self-inactivating invertebrate opsin and its RGS8-fusion protein can function as optical control tools biased for Gβγ-dependent ion channel modulation.
Collapse
Affiliation(s)
- Hisao Tsukamoto
- Department of Biology, Kobe University, Kobe657-8501, Japan
- Department of Life and Coordination-Complex Molecular Science, Institute for Molecular Science, Okazaki444-8585, Japan
- Japan Science and Technology Agency, Precursory Research for Embryonic Science and Technology, Kawaguchi 332-0012, Japan
| | - Yoshihiro Kubo
- Division of Biophysics and Neurobiology, Department of Molecular Physiology, National Institute for Physiological Sciences, Okazaki444-8585, Japan
- Department of Physiological Sciences, The Graduate University for Advanced Studies, Hayama240-0193, Japan
| |
Collapse
|
41
|
Watkins de Jong L, Nejad MM, Yoon E, Cheng S, Diba K. Optogenetics reveals paradoxical network stabilizations in hippocampal CA1 and CA3. Curr Biol 2023; 33:1689-1703.e5. [PMID: 37023753 PMCID: PMC10175182 DOI: 10.1016/j.cub.2023.03.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 02/22/2023] [Accepted: 03/10/2023] [Indexed: 04/08/2023]
Abstract
Recurrent connectivity between excitatory neurons and the strength of feedback from inhibitory neurons are critical determinants of the dynamics and computational properties of neuronal circuits. Toward a better understanding of these circuit properties in regions CA1 and CA3 of the hippocampus, we performed optogenetic manipulations combined with large-scale unit recordings in rats under anesthesia and in quiet waking, using photoinhibition and photoexcitation with different light-sensitive opsins. In both regions, we saw striking paradoxical responses: subsets of cells increased firing during photoinhibition, while other cells decreased firing during photoexcitation. These paradoxical responses were more prominent in CA3 than in CA1, but, notably, CA1 interneurons showed increased firing in response to photoinhibition of CA3. These observations were recapitulated in simulations where we modeled both CA1 and CA3 as inhibition-stabilized networks in which strong recurrent excitation is balanced by feedback inhibition. To directly test the inhibition-stabilized model, we performed large-scale photoinhibition directed at (GAD-Cre) inhibitory cells and found that interneurons in both regions increased firing when photoinhibited, as predicted. Our results highlight the often-paradoxical circuit dynamics that are evidenced during optogenetic manipulations and indicate that, contrary to long-standing dogma, both CA1 and CA3 hippocampal regions display strongly recurrent excitation, which is stabilized through inhibition.
Collapse
Affiliation(s)
- Laurel Watkins de Jong
- Department of Anesthesiology, Michigan Medicine, 1150 W. Medical Center Dr, Ann Arbor, MI 48109, USA; Department of Psychology, University of Wisconsin-Milwaukee, 2441 E Hartford Ave, Milwaukee, WI 53211, USA
| | | | - Euisik Yoon
- Department of Electrical Engineering and Computer Science, 1301 Beal Avenue, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sen Cheng
- Institute for Neural Computation, Ruhr University Bochum, Universitätsstr. 150, 44801 Bochum, Germany
| | - Kamran Diba
- Department of Anesthesiology, Michigan Medicine, 1150 W. Medical Center Dr, Ann Arbor, MI 48109, USA; Department of Psychology, University of Wisconsin-Milwaukee, 2441 E Hartford Ave, Milwaukee, WI 53211, USA; Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
42
|
Porta-de-la-Riva M, Gonzalez AC, Sanfeliu-Cerdán N, Karimi S, Malaiwong N, Pidde A, Morales-Curiel LF, Fernandez P, González-Bolívar S, Hurth C, Krieg M. Neural engineering with photons as synaptic transmitters. Nat Methods 2023; 20:761-769. [PMID: 37024651 DOI: 10.1038/s41592-023-01836-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 03/03/2023] [Indexed: 04/08/2023]
Abstract
Neuronal computation is achieved through connections of individual neurons into a larger network. To expand the repertoire of endogenous cellular communication, we developed a synthetic, photon-assisted synaptic transmission (PhAST) system. PhAST is based on luciferases and channelrhodopsins that enable the transmission of a neuronal state across space, using photons as neurotransmitters. PhAST overcomes synaptic barriers and rescues the behavioral deficit of a glutamate mutant with conditional, calcium-triggered photon emission between two neurons of the Caenorhabditis elegans nociceptive avoidance circuit. To demonstrate versatility and flexibility, we generated de novo synaptic transmission between two unconnected cells in a sexually dimorphic neuronal circuit, suppressed endogenous nocifensive response through activation of an anion channelrhodopsin and switched attractive to aversive behavior in an olfactory circuit. Finally, we applied PhAST to dissect the calcium dynamics of the temporal pattern generator in a motor circuit for ovipositioning. In summary, we established photon-based synaptic transmission that facilitates the modification of animal behavior.
Collapse
Affiliation(s)
| | | | | | - Shadi Karimi
- Institut de Ciències Fotòniques, Castelldefels, Spain
| | | | | | | | | | | | - Cedric Hurth
- Institut de Ciències Fotòniques, Castelldefels, Spain
| | - Michael Krieg
- Institut de Ciències Fotòniques, Castelldefels, Spain.
| |
Collapse
|
43
|
Schulz A, Richter F, Richter A. In vivo optogenetic inhibition of striatal parvalbumin-reactive interneurons induced genotype-specific changes in neuronal activity without dystonic signs in male DYT1 knock-in mice. J Neurosci Res 2023; 101:448-463. [PMID: 36546658 DOI: 10.1002/jnr.25157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 10/30/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022]
Abstract
The pathophysiology of early-onset torsion dystonia (TOR1A/DYT1) remains unclear. Like 70% of human mutation carriers, rodent models with ΔGAG mutation such as DYT1 knock-in (KI) mice do not show overt dystonia but have subtle sensorimotor deficits and pattern of abnormal synaptic plasticity within the striatal microcircuits. There is evidence that dysfunction of striatal parvalbumin-reactive (Parv+) fast-spiking interneurons (FSIs) can be involved in dystonic signs. To elucidate the relevance of these GABAergic interneurons in the pathophysiology of DYT1 dystonia, we used in vivo optogenetics to specifically inhibit Parv+ and to detect changes in motor behavior and neuronal activity. Optogenetic fibers were bilaterally implanted into the dorsal striatum of male DYT1 KI mice and wild-type (WT) littermates expressing halorhodopsin (eNpHR3.0) in Parv+ interneurons. While stimulations with yellow light pulses for up to 60 min at different pulse durations and interval lengths did not induce abnormal movements, such as dystonic signs, immunohistochemical examinations revealed genotype-dependent differences. In contrast to WT mice, stimulated DYT1 KI showed decreased striatal neuronal activity, that is, less c-Fos reactive neurons, and increased activation of cholinergic interneurons after optogenetic inhibition of Parv+ interneurons. These findings suggest an involvement of Parv+ interneurons in an impaired striatal network in DYT1 KI mice, but at least short-term inhibition of these GABAergic interneurons is not sufficient to trigger a dystonic phenotype, similar to previously shown optogenetic activation of cholinergic interneurons.
Collapse
Affiliation(s)
- Anja Schulz
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Franziska Richter
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany.,Institute of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Angelika Richter
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| |
Collapse
|
44
|
Barać U, Perc M, Gosak M. Determinants of collective failure in excitable networks. CHAOS (WOODBURY, N.Y.) 2023; 33:043120. [PMID: 37097938 DOI: 10.1063/5.0149578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 03/27/2023] [Indexed: 06/19/2023]
Abstract
We study collective failures in biologically realistic networks that consist of coupled excitable units. The networks have broad-scale degree distribution, high modularity, and small-world properties, while the excitable dynamics is determined by the paradigmatic FitzHugh-Nagumo model. We consider different coupling strengths, bifurcation distances, and various aging scenarios as potential culprits of collective failure. We find that for intermediate coupling strengths, the network remains globally active the longest if the high-degree nodes are first targets for inactivation. This agrees well with previously published results, which showed that oscillatory networks can be highly fragile to the targeted inactivation of low-degree nodes, especially under weak coupling. However, we also show that the most efficient strategy to enact collective failure does not only non-monotonically depend on the coupling strength, but it also depends on the distance from the bifurcation point to the oscillatory behavior of individual excitable units. Altogether, we provide a comprehensive account of determinants of collective failure in excitable networks, and we hope this will prove useful for better understanding breakdowns in systems that are subject to such dynamics.
Collapse
Affiliation(s)
- Uroš Barać
- Department of Physics, Faculty of Natural Sciences and Mathematics, University of Maribor, Koroška cesta 160, 2000 Maribor, Slovenia
| | - Matjaž Perc
- Department of Physics, Faculty of Natural Sciences and Mathematics, University of Maribor, Koroška cesta 160, 2000 Maribor, Slovenia
- Alma Mater Europaea, Slovenska ulica 17, 2000 Maribor, Slovenia
- Department of Medical Research, China Medical University Hospital, China Medical University, 40447 Taichung, Taiwan
- Complexity Science Hub Vienna, Josefstädterstrasse 39, 1080 Vienna, Austria
- Department of Physics, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, 02447 Seoul, Republic of Korea
| | - Marko Gosak
- Department of Physics, Faculty of Natural Sciences and Mathematics, University of Maribor, Koroška cesta 160, 2000 Maribor, Slovenia
- Alma Mater Europaea, Slovenska ulica 17, 2000 Maribor, Slovenia
- Institute of Physiology, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| |
Collapse
|
45
|
Formozov A, Dieter A, Wiegert JS. A flexible and versatile system for multi-color fiber photometry and optogenetic manipulation. CELL REPORTS METHODS 2023; 3:100418. [PMID: 37056369 PMCID: PMC10088095 DOI: 10.1016/j.crmeth.2023.100418] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 12/20/2022] [Accepted: 02/08/2023] [Indexed: 03/09/2023]
Abstract
Here, we present simultaneous fiber photometry recordings and optogenetic stimulation based on a multimode fused fiber coupler for both light delivery and collection without the need for dichroic beam splitters. In combination with a multi-color light source and appropriate optical filters, our approach offers remarkable flexibility in experimental design and facilitates the exploration of new molecular tools in vivo at minimal cost. We demonstrate straightforward re-configuration of the setup to operate with green, red, and near-infrared calcium indicators with or without simultaneous optogenetic stimulation and further explore the multi-color photometry capabilities of the system. The ease of assembly, operation, characterization, and customization of this platform holds the potential to foster the development of experimental strategies for multi-color fused fiber photometry combined with optogenetics far beyond its current state.
Collapse
Affiliation(s)
- Andrey Formozov
- Research Group Synaptic Wiring and Information Processing, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurophysiology, MCTN, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Alexander Dieter
- Research Group Synaptic Wiring and Information Processing, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurophysiology, MCTN, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - J. Simon Wiegert
- Research Group Synaptic Wiring and Information Processing, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurophysiology, MCTN, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| |
Collapse
|
46
|
Angelucci A, Clark A, Ingold A, Reiche C, Cundy D, Balsor J, Federer F, McAlinden N, Cheng Y, Rolston J, Rieth L, Dawson M, Mathieson K, Blair S. An Optrode Array for Spatiotemporally Precise Large-Scale Optogenetic Stimulation of Deep Cortical Layers in Non-human Primates. RESEARCH SQUARE 2023:rs.3.rs-2322768. [PMID: 36909489 PMCID: PMC10002840 DOI: 10.21203/rs.3.rs-2322768/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Optogenetics has transformed studies of neural circuit function, but remains challenging to apply in non-human primates (NHPs). A major challenge is delivering intense and spatially precise patterned photostimulation across large volumes in deep tissue. Here, we have developed and validated the Utah Optrode Array (UOA) to meet this critical need. The UOA is a 10×10 glass waveguide array bonded to an electrically-addressable μLED array. In vivo electrophysiology and immediate early gene (c-fos) immunohistochemistry demonstrated the UOA allows for large-scale spatiotemporally precise neuromodulation of deep tissue in macaque primary visual cortex. Specifically, the UOA permits both focal (single layers or columns), and large-scale (across multiple layers or columns) photostimulation of deep cortical layers, simply by varying the number of simultaneously activated μLEDs and/or the light irradiance. These results establish the UOA as a powerful tool for studying targeted neural populations within single or across multiple deep layers in complex NHP circuits.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - John Rolston
- Brigham & Women's Hospital and Harvard Medical School
| | | | | | | | | |
Collapse
|
47
|
Bercovici DA, Princz-Lebel O, Schumacher JD, Lo VM, Floresco SB. Temporal Dynamics Underlying Prelimbic Prefrontal Cortical Regulation of Action Selection and Outcome Evaluation during Risk/Reward Decision-Making. J Neurosci 2023; 43:1238-1255. [PMID: 36609453 PMCID: PMC9962784 DOI: 10.1523/jneurosci.0802-22.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 12/23/2022] [Accepted: 12/30/2022] [Indexed: 01/09/2023] Open
Abstract
Risk/reward decision-making is a dynamic process that includes periods of deliberation before action selection and evaluation of the action outcomes that bias subsequent choices. Inactivation of the prelimbic (PL) cortex has revealed its integral role in updating decision biases in the face of changes in probabilistic reward contingencies, yet how phasic PL signals during different phases of the decision process influence choice remains unclear. We used temporally specific optogenetic inhibition to selectively disrupt PL activity coinciding with action selection and outcome phases to examine how these signals influence choice. Male rats expressing the inhibitory opsin eArchT within PL excitatory neurons were well trained on a probabilistic discounting task, entailing choice between small/certain versus large/risky rewards, the probability of which varied over a session (50-12.5%). During testing, brief light pulses suppressed PL activity before choice or after different outcomes. Prechoice suppression reduced bias toward more preferred/higher utility options and disrupted how recent outcomes influenced subsequent choice. Inhibition during risky losses induced a similar profile, but here, the impact of reward omissions were either amplified or diminished, relative to the context of the estimated profitability of the risky option. Inhibition during large or small reward receipt reduced risky choice when this option was more profitable, suggesting these signals can both reinforce rewarded risky choices and also act as a relative value comparator signal that augments incentive for larger rewards. These findings reveal multifaceted contributions by the PL in implementing decisions and integrating action-outcome feedback to assign context to the decision space.SIGNIFICANCE STATEMENT The PL prefrontal cortex plays an integral role in guiding risk/reward decisions, but how activity in this region during different phases of the decision process influences choice is unclear. By using temporally specific optogenetic manipulations of this activity, the present study unveiled previously uncharacterized and differential contributions by PL in implementing decision policies and how evaluation of decision outcomes shape subsequent choice. These findings provide novel insight into the dynamic processes engaged by the PL that underlie action selection in situations involving reward uncertainty that may aid in understanding the mechanism underlying normal and aberrant decision-making processes.
Collapse
Affiliation(s)
- Debra A Bercovici
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T1Z4, Canada
| | - Oren Princz-Lebel
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T1Z4, Canada
| | - Jackson D Schumacher
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T1Z4, Canada
| | - Valerie M Lo
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T1Z4, Canada
| | - Stan B Floresco
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T1Z4, Canada
| |
Collapse
|
48
|
Vettkötter D, Schneider M, Goulden BD, Dill H, Liewald J, Zeiler S, Guldan J, Ateş YA, Watanabe S, Gottschalk A. Rapid and reversible optogenetic silencing of synaptic transmission by clustering of synaptic vesicles. Nat Commun 2022; 13:7827. [PMID: 36535932 PMCID: PMC9763335 DOI: 10.1038/s41467-022-35324-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Acutely silencing specific neurons informs about their functional roles in circuits and behavior. Existing optogenetic silencers include ion pumps, channels, metabotropic receptors, and tools that damage the neurotransmitter release machinery. While the former hyperpolarize the cell, alter ionic gradients or cellular biochemistry, the latter allow only slow recovery, requiring de novo synthesis. Thus, tools combining fast activation and reversibility are needed. Here, we use light-evoked homo-oligomerization of cryptochrome CRY2 to silence synaptic transmission, by clustering synaptic vesicles (SVs). We benchmark this tool, optoSynC, in Caenorhabditis elegans, zebrafish, and murine hippocampal neurons. optoSynC clusters SVs, observable by electron microscopy. Locomotion silencing occurs with tauon ~7.2 s and recovers with tauoff ~6.5 min after light-off. optoSynC can inhibit exocytosis for several hours, at very low light intensities, does not affect ion currents, biochemistry or synaptic proteins, and may further allow manipulating different SV pools and the transfer of SVs between them.
Collapse
Affiliation(s)
- Dennis Vettkötter
- Buchmann Institute for Molecular Life Sciences, Goethe University, D-60438, Frankfurt, Germany
- Institute of Biophysical Chemistry, Goethe University, D-60438, Frankfurt, Germany
| | - Martin Schneider
- Buchmann Institute for Molecular Life Sciences, Goethe University, D-60438, Frankfurt, Germany
- Institute of Biophysical Chemistry, Goethe University, D-60438, Frankfurt, Germany
- Max Planck Institute for Neurobiology, D-82152, Martinsried, Germany
| | - Brady D Goulden
- Department of Cell Biology and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Holger Dill
- Buchmann Institute for Molecular Life Sciences, Goethe University, D-60438, Frankfurt, Germany
- Institute of Biophysical Chemistry, Goethe University, D-60438, Frankfurt, Germany
| | - Jana Liewald
- Buchmann Institute for Molecular Life Sciences, Goethe University, D-60438, Frankfurt, Germany
- Institute of Biophysical Chemistry, Goethe University, D-60438, Frankfurt, Germany
| | - Sandra Zeiler
- Buchmann Institute for Molecular Life Sciences, Goethe University, D-60438, Frankfurt, Germany
- Institute of Biophysical Chemistry, Goethe University, D-60438, Frankfurt, Germany
| | - Julia Guldan
- Buchmann Institute for Molecular Life Sciences, Goethe University, D-60438, Frankfurt, Germany
- Master Program Interdisciplinary Neurosciences, Department of Biological Sciences, Goethe University, Frankfurt, Germany
| | - Yilmaz Arda Ateş
- Buchmann Institute for Molecular Life Sciences, Goethe University, D-60438, Frankfurt, Germany
- Master Program Interdisciplinary Neurosciences, Department of Biological Sciences, Goethe University, Frankfurt, Germany
| | - Shigeki Watanabe
- Department of Cell Biology and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Alexander Gottschalk
- Buchmann Institute for Molecular Life Sciences, Goethe University, D-60438, Frankfurt, Germany.
- Institute of Biophysical Chemistry, Goethe University, D-60438, Frankfurt, Germany.
| |
Collapse
|
49
|
Temporal dynamics of the sensorimotor convergence underlying voluntary limb movement. Proc Natl Acad Sci U S A 2022; 119:e2208353119. [PMID: 36409890 PMCID: PMC9860324 DOI: 10.1073/pnas.2208353119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Descending motor drive and somatosensory feedback play important roles in modulating muscle activity. Numerous studies have characterized the organization of neuronal connectivity in which descending motor pathways and somatosensory afferents converge on spinal motor neurons as a final common pathway. However, how inputs from these two pathways are integrated into spinal motor neurons to generate muscle activity during actual motor behavior is unknown. Here, we simultaneously recorded activity in the motor cortices (MCx), somatosensory afferent neurons, and forelimb muscles in monkeys performing reaching and grasping movements. We constructed a linear model to explain the instantaneous muscle activity using the activity of MCx (descending input) and peripheral afferents (afferent input). Decomposition of the reconstructed muscle activity into each subcomponent indicated that muscle activity before movement onset could first be explained by descending input from mainly the primary motor cortex and muscle activity after movement onset by both descending and afferent inputs. Descending input had a facilitative effect on all muscles, whereas afferent input had a facilitative or suppressive effect on each muscle. Such antagonistic effects of afferent input can be explained by reciprocal effects of the spinal reflex. These results suggest that descending input contributes to the initiation of limb movement, and this initial movement subsequently affects muscle activity via the spinal reflex in conjunction with the continuous descending input. Thus, spinal motor neurons are subjected to temporally organized modulation by direct activation through the descending pathway and the lagged action of the spinal reflex during voluntary limb movement.
Collapse
|
50
|
Lee JY, You T, Woo CW, Kim SG. Optogenetic fMRI for Brain-Wide Circuit Analysis of Sensory Processing. Int J Mol Sci 2022; 23:ijms232012268. [PMID: 36293125 PMCID: PMC9602603 DOI: 10.3390/ijms232012268] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/12/2022] [Accepted: 10/12/2022] [Indexed: 11/20/2022] Open
Abstract
Sensory processing is a complex neurological process that receives, integrates, and responds to information from one's own body and environment, which is closely related to survival as well as neurological disorders. Brain-wide networks of sensory processing are difficult to investigate due to their dynamic regulation by multiple brain circuits. Optogenetics, a neuromodulation technique that uses light-sensitive proteins, can be combined with functional magnetic resonance imaging (ofMRI) to measure whole-brain activity. Since ofMRI has increasingly been used for investigating brain circuits underlying sensory processing for over a decade, we systematically reviewed recent ofMRI studies of sensory circuits and discussed the challenges of optogenetic fMRI in rodents.
Collapse
Affiliation(s)
- Jeong-Yun Lee
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, Korea
| | - Taeyi You
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, Korea
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Korea
| | - Choong-Wan Woo
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, Korea
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Korea
| | - Seong-Gi Kim
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon 16419, Korea
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Korea
- Correspondence: ; Tel.: +82-31-299-4350; Fax: +82-31-299-4506
| |
Collapse
|